US6767544B2 - Methods for treating cardiovascular diseases with botulinum toxin - Google Patents

Methods for treating cardiovascular diseases with botulinum toxin Download PDF

Info

Publication number
US6767544B2
US6767544B2 US10/114,740 US11474002A US6767544B2 US 6767544 B2 US6767544 B2 US 6767544B2 US 11474002 A US11474002 A US 11474002A US 6767544 B2 US6767544 B2 US 6767544B2
Authority
US
United States
Prior art keywords
blood vessel
botulinum toxin
toxin
botulinum
botulinum neurotoxin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US10/114,740
Other versions
US20030185860A1 (en
Inventor
Gregory F. Brooks
Stephen Donovan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Allergan Inc
Original Assignee
Allergan Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=28453838&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US6767544(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Allergan Inc filed Critical Allergan Inc
Priority to US10/114,740 priority Critical patent/US6767544B2/en
Assigned to ALLERGAN SALES, INC. reassignment ALLERGAN SALES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BROOKS, GREGORY F.
Priority to EP03716821A priority patent/EP1490097B2/en
Priority to AT03716821T priority patent/ATE355852T1/en
Priority to JP2003581806A priority patent/JP2005521735A/en
Priority to AU2003220511A priority patent/AU2003220511C1/en
Priority to KR10-2004-7015481A priority patent/KR20040105818A/en
Priority to MXPA04009410A priority patent/MXPA04009410A/en
Priority to ES03716821T priority patent/ES2281636T5/en
Priority to CA002480505A priority patent/CA2480505A1/en
Priority to DE60312309T priority patent/DE60312309T3/en
Priority to BR0308928-2A priority patent/BR0308928A/en
Priority to PCT/US2003/009157 priority patent/WO2003084567A1/en
Assigned to ALLERGAN, INC. reassignment ALLERGAN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DONOVAN, STEPHEN
Assigned to ALLERGAN, INC. reassignment ALLERGAN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ALLERGAN SALES, INC. (MERGED INTO ALLERGAN SALES, LLC 6/3/2002)
Priority to US10/628,905 priority patent/US7048927B2/en
Publication of US20030185860A1 publication Critical patent/US20030185860A1/en
Priority to US10/870,603 priority patent/US7494654B2/en
Publication of US6767544B2 publication Critical patent/US6767544B2/en
Application granted granted Critical
Priority to HK05103242A priority patent/HK1070575A1/en
Priority to US11/345,736 priority patent/US7223399B2/en
Priority to US11/446,403 priority patent/US20060216313A1/en
Priority to JP2009032291A priority patent/JP2009102440A/en
Priority to US12/436,730 priority patent/US20090220568A1/en
Priority to JP2012277214A priority patent/JP2013053167A/en
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/04Macromolecular materials
    • A61L31/043Proteins; Polypeptides; Degradation products thereof
    • A61L31/047Other specific proteins or polypeptides not covered by A61L31/044 - A61L31/046
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • A61K38/4893Botulinum neurotoxin (3.4.24.69)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/08Clostridium, e.g. Clostridium tetani
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to methods of preventing or reducing restenosis that may occur in blood vessels after mechanically expanding the diameter of an occluded blood vessel.
  • Atherosclerosis is a progressive disease wherein fatty, fibrous, calcific, or thrombotic deposits produce atheromatous plaques, within and beneath the intima which is the innermost layer of arteries.
  • Atherosclerosis tends to involve large and medium sized arteries. The most commonly affected are the aorta, iliac, femoral, coronary, and cerebral arteries. Clinical symptoms occur because the mass of the atherosclerotic plaque reduces blood flow through the afflicted artery, thereby compromising tissue or organ function distal to it.
  • Percutaneous transluminal coronary angioplasty is a non-surgical method for treatment of coronary atherosclerosis.
  • an inflatable balloon is inserted in a coronary artery in the region of arterial narrowing. Inflation of the balloon for 15-30 seconds results in an expansion of the narrowed lumen or passageway. Because residual narrowing is usually present after the first balloon inflation, multiple or prolonged inflations are routinely performed to reduce the severity of the residual tube narrowing.
  • Stents are often used in combination with coronary balloon angioplasty.
  • a stent is used to brace the blood vessel open after an initial expansion of the narrowed blood vessel by a balloon.
  • Self expanding stents are also used to expand and hold open occluded blood vessels.
  • Various stents and their use are disclosed in U.S. Pat. Nos. 6,190,404; 6,344,055; 6,306,162; 6,293,959; 6,270,521; 6,264,671; 6,261,318; 6,241,758; 6,217,608; 6,196,230; 6,183,506; 5,989,280. The disclosure of each of these patents is incorporated in its entirety herein by reference.
  • angioplasty One problem with angioplasty is that following the procedure restenosis, or recurrence of the obstruction, may occur. Tears in the wall expose blood to foreign material and proteins, such as collagen, which are highly thrombogenic. Resulting clots can contain growth hormones which may be released by platelets within the clot. Additionally, thrombosis may cause release of growth hormones and cytokines by cells from macrophages. Growth hormones may cause smooth muscle cells and fibroblasts to aggregate in the region and multiply. Further, following angioplasty there is often a loss of the single layer of cells that normally covers the internal surface of blood vessels which leads to thrombosis.
  • Angioplasty procedures also produce injuries in the arterial wall which become associated with inflammation. Any kind of inflammatory response may cause growth of new tissue, for example, scar tissue, which may contribute to restenosis.
  • One of the other major causes of restenosis following angioplasty may be that the injured arterial wall may exhibit a reduced hemocompatability compared to that associated with a normal arterial wall.
  • Adverse responses which are associated with reduced hemocompatability include platelet adhesion, aggregation, and activation; thrombosis; inflammatory cell reactions such as adhesion and activation of monocytes or macrophages; and the infiltration of leukocytes into the arterial wall.
  • Restenosis is a serious problem that may occur in over one third of all coronary angioplasty patients. Therefore, there exists a need for methods to reduce or eliminate the occurrence of restenosis which may follow procedures to mechanically expand an occluded blood vessel.
  • Clostridium botulinum produces a potent polypeptide neurotoxin, botulinum toxin, which causes a neuroparalytic illness in humans and animals referred to as botulism.
  • the spores of Clostridium botulinum are found in soil and can grow in improperly sterilized and sealed food containers of home based canneries, which are the cause of many of the cases of botulism.
  • the effects of botulism typically appear 18 to 36 hours after eating the foodstuffs infected with a Clostridium botulinum culture or spores.
  • the botulinum toxin can apparently pass unattenuated through the lining of the gut and attack peripheral motor neurons. Symptoms of botulinum toxin intoxication can progress from difficulty walking, swallowing, and speaking to paralysis of the respiratory muscles and death.
  • Botulinum toxin type A (“BoNT/A”) is the most lethal natural biological agent known to man. About 50 picograms of botulinum toxin (purified neurotoxin complex) serotype A is a LD 50 in mice. One unit (U) of botulinum toxin is defined as the LD 50 upon intraperitoneal injection into female Swiss Webster mice weighing 18-20 grams each. Seven immunologically distinct botulinum neurotoxins have been characterized, these being respectively botulinum neurotoxin serotypes A, B, C 1 , D, E, F and G each of which is distinguished by neutralization with serotype-specific antibodies.
  • botulinum toxin serotype B botulinum toxin serotype B
  • BoNt/A botulinum toxin serotype B
  • BoNt/B botulinum toxin type B
  • Botulinum toxin apparently binds with high affinity to cholinergic motor neurons, is translocated into the neuron and blocks the release of acetylcholine.
  • Botulinum toxins have been used in clinical settings for the treatment of neuromuscular disorders characterized by hyperactive skeletal muscles.
  • BoNt/A has been approved by the U.S. Food and Drug Administration for the treatment of blepharospasm, strabismus, hemifacial spasm and cervical dystonia. Additionally, a botulinum toxin type B has been approved by the FDA for the treatment of cervical dystonia.
  • Non-serotype A botulinum toxin serotypes apparently have a lower potency and/or a shorter duration of activity as compared to BoNt/A.
  • Clinical effects of peripheral intramuscular BoNt/A are usually seen within one week of injection. The typical duration of symptomatic relief from a single intramuscular injection of BoNt/A averages about three months.
  • botulinum toxins serotypes Although all the botulinum toxins serotypes apparently inhibit release of the neurotransmitter acetylcholine at the neuromuscular junction, they do so by affecting different neurosecretory proteins and/or cleaving these proteins at different sites.
  • botulinum serotypes A and E both cleave the 25 kiloDalton (kD) synaptosomal associated protein (SNAP-25), but they target different amino acid sequences within this protein.
  • BoNT/B, D, F and G act on vesicle-associated protein (VAMP, also called synaptobrevin), with each serotype cleaving the protein at a different site.
  • VAMP vesicle-associated protein
  • botulinum toxin serotype C 1 (BoNT/C 1 ) has been shown to cleave both syntaxin and SNAP-25. These differences in mechanism of action may affect the relative potency and/or duration of action of the various botulinum toxin serotypes.
  • the molecular mechanism of toxin intoxication appears to be similar and to involve at least three steps or stages.
  • the toxin binds to the presynaptic membrane of the target neuron through a specific interaction between the H chain and a cell surface receptor; the receptor is thought to be different for each serotype of botulinum toxin and for tetanus toxin.
  • the carboxyl end segment of the H chain, H C appears to be important for targeting of the toxin to the cell surface.
  • the toxin crosses the plasma membrane of the poisoned cell.
  • the toxin is first engulfed by the cell through receptor-mediated endocytosis, and an endosome containing the toxin is formed.
  • the toxin escapes the endosome into the cytoplasm of the cell.
  • This last step is thought to be mediated by the amino end segment of the H chain, H N , which triggers a conformational change of the toxin in response to a pH of about 5.5 or lower.
  • Endosomes are known to possess a proton pump which decreases intra endosomal pH.
  • the conformational shift exposes hydrophobic residues in the toxin, which permits the toxin to embed itself in the endosomal membrane.
  • the toxin then translocates through the endosomal membrane into the cytosol.
  • the last step of the mechanism of botulinum toxin activity appears to involve reduction of the disulfide bond joining the H and L chain.
  • the entire toxic activity of botulinum and tetanus toxins is contained in the L chain of the holotoxin; the L chain is a zinc (Zn++) endopeptidase which selectively cleaves proteins essential for recognition and docking of neurotransmitter-containing vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane.
  • VAMP vesicle-associated membrane protein
  • Each toxin specifically cleaves a different bond.
  • the botulinum toxins are released by Clostridial bacterium as complexes comprising the 150 kD botulinum toxin protein molecule along with associated non-toxin proteins.
  • the BoNt/A complex can be produced by Clostridial bacterium as 900 kD, 500 kD and 300 kD forms.
  • BoNT/B and C 1 are apparently produced as only a 500 kD complex.
  • BoNT/D is produced as both 300 kD and 500 kD complexes.
  • BoNT/E and F are produced as only approximately 300 kD complexes.
  • the complexes i.e. molecular weight greater than about 150 kD
  • These two non-toxin proteins may act to provide stability against denaturation to the botulinum toxin molecule and protection against digestive acids when toxin is ingested.
  • botulinum toxin complexes may result in a slower rate of diffusion of the botulinum toxin away from a site of intramuscular injection of a botulinum toxin complex.
  • botulinum toxin inhibits potassium cation induced release of both acetylcholine and norepinephrine from primary cell cultures of brainstem tissue. Additionally, it has been reported that botulinum toxin inhibits the evoked release of both glycine and glutamate in primary cultures of spinal cord neurons and that in brain synaptosome preparations botulinum toxin inhibits the release of each of the neurotransmitters acetylcholine, dopamine, norepinephrine, CGRP and glutamate.
  • BoNt/A can be obtained by establishing and growing cultures of Clostridium botulinum in a fermenter and then harvesting and purifying the fermented mixture in accordance with known procedures. All the botulinum toxin serotypes are initially synthesized as inactive single chain proteins which must be cleaved or nicked by proteases to become neuroactive. The bacterial strains that make botulinum toxin serotypes A and G possess endogenous proteases and serotypes A and G can therefore be recovered from bacterial cultures in predominantly their active form. In contrast, botulinum toxin serotypes C 1 , D and E are synthesized by nonproteolytic strains and are therefore typically unactivated when recovered from culture.
  • Serotypes B and F are produced by both proteolytic and nonproteolytic strains and therefore can be recovered in either the active or inactive form.
  • the proteolytic strains that produce, for example, the BoNt/B serotype only cleave a portion of the toxin produced.
  • the exact proportion of nicked to unnicked molecules depends on the length of incubation and the temperature of the culture. Therefore, a certain percentage of any preparation of, for example, the BoNt/B toxin is likely to be inactive, possibly accounting for the known significantly lower potency of BoNt/B as compared to BoNt/A.
  • BoNt/B has, upon intramuscular injection, a shorter duration of activity and is also less potent than BoNt/A at the same dose level.
  • BoNt/A has been used clinically as follows:
  • extraocular muscles have been injected intramuscularly with between about 1-5 units of BOTOX®, the amount injected varying based upon both the size of the muscle to be injected and the extent of muscle paralysis desired (i.e. amount of diopter correction desired).
  • biceps brachii 50 U to 200 U.
  • Each of the five indicated muscles has been injected at the same treatment session, so that the patient receives from 90 U to 360 U of upper limb flexor muscle BOTOX® by intramuscular injection at each treatment session.
  • the tetanus neurotoxin acts mainly in the central nervous system, while botulinum neurotoxin acts at the neuromuscular junction; both act by inhibiting acetylcholine release from the axon of the affected neuron into the synapse, resulting in paralysis.
  • the effect of intoxication on the affected neuron is long-lasting and until recently has been thought to be irreversible.
  • the tetanus neurotoxin is known to exist in one immunologically distinct serotype.
  • the neurotransmitter acetylcholine is secreted by neurons in many areas of the brain, but specifically by the large pyramidal cells of the motor cortex, by several different neurons in the basal ganglia, by the motor neurons that innervate the skeletal muscles, by the preganglionic neurons of the autonomic nervous system (both sympathetic and parasympathetic), by the postganglionic neurons of the parasympathetic nervous system, and by some of the postganglionic neurons of the sympathetic nervous system.
  • acetylcholine has an excitatory effect.
  • acetylcholine is known to have inhibitory effects at some of the peripheral parasympathetic nerve endings, such as inhibition of the heart by the vagal nerve.
  • the efferent signals of the autonomic nervous system are transmitted to the body through either the sympathetic nervous system or the parasympathetic nervous system.
  • the preganglionic neurons of the sympathetic nervous system extend from preganglionic sympathetic neuron cell bodies located in the intermediolateral horn of the spinal cord.
  • the preganglionic sympathetic nerve fibers, extending from the cell body synapse with postganglionic neurons located in either a paravertebral sympathetic ganglion or in a prevertebral ganglion. Since, the preganglionic neurons of both the sympathetic and parasympathetic nervous system are cholinergic, application of acetylcholine to the ganglia will excite both sympathetic and parasympathetic postganglionic neurons.
  • Acetylcholine activates two types of receptors, muscarinic and nicotinic receptors.
  • the muscarinic receptors are found in all effector cells stimulated by the postganglionic neurons of the parasympathetic nervous system, as well as in those stimulated by the postganglionic cholinergic neurons of the sympathetic nervous system.
  • the nicotinic receptors are found in the synapses between the preganglionic and postganglionic neurons of both the sympathetic and parasympathetic.
  • the nicotinic receptors are also present in many membranes of skeletal muscle fibers at the neuromuscular junction.
  • Acetylcholine is released from cholinergic neurons when small, clear, intracellular vesicles fuse with the presynaptic neuronal cell membrane.
  • a wide variety of non-neuronal secretory cells such as, adrenal medulla (as well as the PC12 cell line) and pancreatic islet cells release catecholamines and insulin, respectively, from large dense-core vesicles.
  • the PC12 cell line is a clone of rat pheochromocytoma cells extensively used as a tissue culture model for studies of sympathoadrenal development.
  • Botulinum toxin inhibits the release of both types of compounds from both types of cells in vitro, permeabilized (as by electroporation) or by direct injection of the toxin into the denervated cell. Botulinum toxin is also known to block release of the neurotransmitter glutamate from cortical synaptosomes cell cultures.
  • the present invention provides for methods to treat cardiovascular diseases in a mammal, for example, in a human.
  • the methods include a step of administering an effective amount of a botulinum toxin directly to a blood vessel of a mammal to treat a cardiovascular disease.
  • treating the cardiovascular disease prevents restenosis.
  • the mammal is having or has had a cardiovascular procedure.
  • the cardiovascular procedure is an arterial cardiovascular procedure, for example, a coronary arterial cardiovascular procedure.
  • the cardiovascular procedure includes an angioplasty procedure.
  • the angioplasty includes the step of inserting a stent into the blood vessel of the mammal. In another embodiment, the angioplasty does not include the step of inserting a stent into a blood vessel.
  • the angioplasty procedure may be, for example, balloon angioplasty.
  • the balloon angioplasty includes the use of a stent. For example, a stent may be inserted into the blood vessel during the balloon angioplasty.
  • the procedure is not limited to use of a balloon. Any device that may be used to mechanically open a constricted blood vessel, for example, a spring or other expanding device, may be used to perform an angioplasty.
  • the step of administering the botulinum toxin may include a step of injecting the botulinum toxin into a wall of the blood vessel.
  • the toxin may be injected into the intima, media and/or adventia layers of the blood vessel.
  • the step of administering may be accomplished using a stent which has been coated or impregnated with botulinum toxin.
  • the botulinum toxin reduces or eliminates damage to a blood vessel. Examples of damage that may occur are stretching and/or tearing of a blood vessel or any other damage that may occur to the blood vessel as a result of mechanically expanding the inner diameter of the blood vessel.
  • the botulinum toxin reduces or eliminates damage to the blood vessel, at least in part, by dilating the blood vessel.
  • the botulinum toxin reduces or eliminates damage to the blood vessel, at least in part, by reducing or eliminating inflammation of the blood vessel.
  • the botulinum toxin may be any botulinum toxin including botulinum toxin types A, B, C, D, E, F, G or mixtures thereof or combinations thereof, including a modified, hybrid or chimeric botulinum toxin.
  • the method includes a step of administering to the mammal an effective amount of botulinum toxin thereby preventing restenosis in a blood vessel.
  • the method includes the step of administering to a mammal an effective amount of a botulinum toxin thereby preventing damage in the blood vessel and preventing restenosis.
  • the present invention also provides for methods to prevent restenosis in a mammal by preventing inflammation in a blood vessel which may occur during or following a cardiovascular procedure.
  • the method includes the step of administering to a mammal an effective amount of the botulinum toxin thereby preventing inflammation in the blood vessel and preventing restenosis.
  • the present invention provides for methods to prevent restenosis in a mammal by dilating a blood vessel proceeding, during or following a cardiovascular procedure.
  • the method includes the step of administering to a mammal an effective amount of the botulinum toxin thereby dilating the blood vessel and preventing restenosis.
  • compositions for use in cardiovascular procedures include a stent with a botulinum toxin attached to the stent or imbedded in the stent.
  • the botulinum toxin may be any botulinum toxin including botulinum toxin type A, B, C, D, E, F, G or combinations thereof or mixtures thereof.
  • Agent is defined as a neurotoxin, for example, a botulinum toxin, for use in accordance with the present invention.
  • An agent may be a fragment of a neurotoxin, a modified neurotoxin or a variant neurotoxin that possesses some or all of the biological activity of an unmodified neurotoxin.
  • Angioplasty means any procedure where the inner diameter of a blood vessel is mechanically expanded.
  • a “botulinum toxin” may refer to native botulinum toxin or a functional fragment of a botulinum toxin or a modified botulinum toxin.
  • botulinum toxins with amino acid deletions, additions, alterations or substitutions that delete, add, alter or substitute a single amino acid, or a small percentage of amino acids are conservatively modified variations of botulinum toxins. Where one or more substitutions of an amino acid(s) with a chemically similar amino acid are made in a botulinum toxin, this also results in a conservatively modified variation of a botulinum toxin.
  • Cardiovascular means pertaining to blood vessels, for example, blood vessels of the heart.
  • Clostidial toxin or “Clostridial neurotoxin” means a toxin produced naturally by the genus of bacteria Clostridium.
  • Clostidial toxins include, but are not limited to, botulinum toxins, tetanus toxins, difficile toxins and butyricum toxins.
  • a Clostridial toxin can also be made by known recombinant means by a non- Clostridial bacterium.
  • “Combination” means an ordered sequence of elements.
  • a combination of botulinum toxins may mean administration of botulinum toxin E, followed by administration of botulinum toxin type A, followed by administration of botulinum toxin type B. This is opposed to a “mixture” where, for example, different toxin types are combined prior to administration.
  • “Damage” means tearing, scratching, stretching, scraping, bruising and/or inflammation or injury caused by inflammation or other injury that may occur in a blood vessel undergoing a procedure, for example, a procedure where the inner diameter of the blood vessel is expanded using mechanical force.
  • “Fragment” means an amino acid sequence that comprises five amino acids or more of the native amino acid sequence up to a size of minus at least one amino acid from the native sequence.
  • a fragment of a botulinum toxin type A light chain comprises five or more amino acids of the amino acid sequence of the native botulinum toxin type A light chain up to a size of minus one amino acid from the native light chain.
  • H C means a fragment obtained from the H chain of a Clostridial toxin which is equivalent, for example functionally equivalent, to the carboxyl end fragment of the H chain, or the portion corresponding to that fragment in the intact H chain involved in binding to a cell surface or cell surface receptor.
  • H N means a fragment or variant obtained from an H chain of a Clostridial toxin which may be functionally equivalent to the portion of an intact H chain involved in the translocation of at least the L chain across an intracellular endosomal membrane into a cytoplasm of a cell.
  • An H N may result from an H C being removed from an H chain.
  • An H N may also result from an H chain being modified such that its H C no longer binds to cholinergic cell surfaces.
  • Heavy chain means the heavy chain of a Clostridial neurotoxin or a fragment or variant of an H N of a Clostridial neurotoxin.
  • a heavy chain may have a molecular weight of about 100 kD and can be referred to as H chain, or as H.
  • LH N means a fragment obtained from a Clostridial neurotoxin that contains the L chain coupled to an H N .
  • LH N can be obtained from the intact Clostridial neurotoxin by proteolysis, so as to remove or to modify the H C domain.
  • Light chain means the light chain of a Clostridial neurotoxin or a fragment or variant of a light chain of a Clostridial neurotoxin.
  • a light chain may have a molecular weight of about 50 kD, and can be referred to as L chain, L, or as the proteolytic domain of a Clostridial neurotoxin.
  • Linker means a molecule which couples two or more other molecules or components together.
  • a “modified neurotoxin” means a neurotoxin that has a nonnative component covalently attached to the neurotoxin and/or a native portion of the neurotoxin missing.
  • a modified botulinum toxin may be a light chain of a botulinum toxin with a substance P molecule covalently attached.
  • Neuron or “toxin” means a substance that inhibits neuronal function or cellular secretion. Clostridial toxins are examples of a neurotoxin.
  • Prevent means to keep from occurring in whole or in part.
  • Reduce means to make smaller in magnitude (e.g. size, quantity or number).
  • the reduction may be about 1% to about 100%.
  • the reduction may be between about 1% and about 10% or between about 10% and about 20% or between about 10% and about 30% or between about 10% and about 40% or between about 10% and about 50% or between about 10% and about 60% or between about 10% and about 70% or between about 10% and about 80% or between about 10% and about 90% or between about 10% and about 100%.
  • Spacer means a molecule or set of molecules which physically separate and/or add distance between components of agents for use in accordance with the invention.
  • substantially means largely but not entirely. For example, substantially may mean about 10% to about 99.999%, about 20% to about 99.999%, about 30% to about 99.999%, about 40% to about 99.999% or about 50% to about 99.999%.
  • Targeting component means a molecule that has a specific binding affinity for a cell surface or cell surface receptor.
  • Variant means a molecule or peptide which is substantially the same as that of a disclosed molecule or peptide in its structure and function.
  • a variant of a specified light chain may have amino acid sequence differences when compared to the amino acid sequence of the specified light chain.
  • Variants may be considered to be equivalent to the specifically disclosed molecules and as such are within the scope of the invention.
  • the present invention is, in part, based upon the discovery that a neurotoxin, for example, botulinum toxin, is useful for treating cardiovascular disease, for example, treating cardiovascular disease in a patient who has undergone, or is undergoing, a cardiovascular procedure.
  • a neurotoxin for example, botulinum toxin
  • the present invention provides for methods to reduce or eliminate restenosis following a cardiovascular procedure.
  • any blood vessel in the body including, but not limited to, coronary (heart), cerebral (brain), Carotid (neck), Renal (kidney), Visceral (abdominal), Iliac (hip), Femoropopliteal (thigh), Infrapopliteal (knee) blood vessels.
  • the invention comprises application of a neurotoxin, for example, a botulinum toxin, to a blood vessel of a patient who is undergoing or will undergo or has undergone a procedure that may, directly or indirectly, lead to damage of a blood vessel, for example, a coronary artery.
  • a neurotoxin for example, a botulinum toxin
  • the present invention provides for methods to treat a patient undergoing an angioplasty procedure such that restenosis is reduced or eliminated following the procedure.
  • the angioplasty includes use of a stent, for example, a self expanding stent.
  • the angioplasty is balloon angioplasty.
  • the angioplasty is balloon angioplasty that includes use of a stent.
  • the angioplasty is balloon angioplasty that does not include use of a stent.
  • the present methods prevent damage to a blood vessel which may occur in association with a mechanical expanding of an otherwise occluded or partially occluded blood vessel. Therefore, methods of the present invention may prevent restenosis that may otherwise occur as a result of such damage. Examples of damage that may be prevented are tearing, scratching, stretching, scraping, bruising and/or inflammation or injury caused by inflammation or other injury that may occur in a blood vessel undergoing a procedure, for example, a procedure where the inner diameter of the blood vessel is expanded using mechanical force.
  • botulinum toxin in preventing damage from occurring in a blood vessel is not completely understood, without wishing to limit the invention to any particular theory or mechanism of operation, the inventor suggests at least two possible theories of operation.
  • the toxins are thought exert a dilating effect on blood vessels thereby increasing the diameter of a vessel, including the inner diameter of the vessel.
  • Optical coherence topography may be used to provide a measure of the dilating effect of the toxin.
  • the dilating effect of the toxin may be quantitated as a factor of the original size of the blood vessel opening before administration of the toxin.
  • the blood vessel opening may be dilated to between about 1.5 ⁇ and about 100 ⁇ the size of the opening before administration of the toxin.
  • the blood vessel opening may be dilated to between about 2 ⁇ and about 5 ⁇ the size of the opening before administration of the toxin.
  • the blood vessel opening may be dilated to between about 2 ⁇ and about 10 ⁇ the size of the opening before administration of the toxin. In another example the blood vessel opening may be dilated to between about 2 ⁇ and about 30 ⁇ the size of the opening before administration of the toxin. In another example the blood vessel opening may be dilated to between about 2 ⁇ and about 50 ⁇ the size of the opening before administration of the toxin. In another example the blood vessel opening may be dilated to between about 50 ⁇ and about 100 ⁇ the size of the opening before administration of the toxin.
  • the dilating of the blood vessels may make the vessels more receptive to intervention procedures. For example, balloon angioplasty and/or insertion of a stent or other mechanical intervention may be less likely to damage the blood vessel when the blood vessel is in the dilated state.
  • the blood vessel may be allowed to dilate before the procedure, for example, an angioplasty procedure, is performed. Whether dilation has taken place, and to what extent dilation has taken place can be determined by a physician of ordinary skill. For example, optical coherence topography may be used to make these determinations.
  • the toxins disclosed herein act on inflammation mediating cells, for example, blood vessel endothelial cells.
  • These cells present many biologically active inflammation mediators which may include bradykinin, nitric oxide and vasoactive intestinal peptide. Release of these and/or other mediators may contribute to the events which cause blood vessel inflammation which may contribute to restenosis.
  • the mediators may be included in vesicles which fuse to the inner surface of the cell membrane thereby releasing the vesicle contents to the outside of the cell. It is theorized that interference with the exocytosis process may be the mode of action of the Clostridial toxins.
  • Clostridial toxins may operate by preventing or reducing the secretion of inflammation producing molecules in blood vessel cells or other cells by cleaving or by otherwise interfering with the function of proteins involved in the secretory process by use of a light chain component, for example, a botulinum light chain component.
  • a heavy chain component for example H N
  • H N may also function in certain embodiments of the present invention by, for example, assisting in the release of an agent of the invention from intracellular vesicles, for example, endosomes.
  • inflammation may either directly, or indirectly contribute to restenosis.
  • blood vessel inflammation that may be associated with cardiovascular procedures, for example, balloon angioplasty and/or insertion of a stent, restenosis may be reduced in a patient who has undergone a cardiovascular procedure.
  • Another possible mechanism for the efficacy of the present disclosed invention is an effect of a botulinum toxin to inhibit neuronally mediated blood vessel contraction.
  • Pretreatment with a botulinum toxin can inhibit a post stretch constriction.
  • a botulinum toxin which is a targeted toxin wherein the native binding moiety of the toxin has been replaced in whole or in part by a new binding moiety which targets the toxin to alpha2 receptors on sympathetic neurons which innervate the blood vessel to be treated.
  • NO can be induced locally to cause dilation.
  • the neurotoxin for use in accordance with the present invention may comprise a targeting component, a therapeutic component and a translocation component.
  • the targeting component comprises a carboxyl end fragment of a heavy chain of a butyricum toxin, a tetani toxin or a botulinum toxin including botulinum toxin types A, B, C, D, E, F and G.
  • the targeting component may be of non-botulinum toxin origin.
  • targeting components that may be used in the present invention include, but are not limited to, antibodies, monoclonal antibodies, antibody fragments (Fab, F(ab)′ 2 , Fv, ScFv, and other antibody fragments of the like), lectins, hormones, cytokines, growth factors, peptides, carbohydrates, lipids, glycons and nucleic acids.
  • Other targeting components that may be useful in accordance with the present invention are disclosed in WO 01/21213 which is incorporated in its entirety herein by reference.
  • One exemplary targeting component for use in accordance with the present invention is substance P or substances similar to substance P.
  • Use of substance P, or substances similar to substance P, as targeting components is described in U.S. patent application Ser. Nos. 09/489,667; 09/922,093 and 09/625,098 each of which is incorporated in its entirety herein by reference.
  • the therapeutic component operates to selectively cleave proteins essential for recognition and docking of secretory vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane.
  • One effect of the therapeutic component may be to substantially interfere with the release of neurotransmitters from a cell.
  • Another effect of the therapeutic component may be to cause dilation of blood vessels.
  • Another effect may be to cause flaccid paralysis of smooth muscle tissue.
  • Another effect may be to reduce or eliminate secretion from cells, for example, inflammation producing cells.
  • the therapeutic component comprises a light chain of a butyricum toxin, a tetani toxin, a botulinum toxin, for example, botulinum toxin type A, B, C, D, E, F and G.
  • the translocation component may facilitate the transfer of at least a part of the neurotoxin, for example the therapeutic component into the cytoplasm of the target cell.
  • the translocation component comprises an amino end fragment of a heavy chain of a butyricum toxin, a tetani toxin, a botulinum toxin, for example, botulinum toxin type A, B, C, D, E, F and G.
  • recombinant DNA methodologies may be used to produce the components of agents useful in accordance with the invention. These techniques may include steps of obtaining cloned genes from natural sources, or from synthetic oligonucleotide sequences, which may encode botulinum neurotoxin components including botulinum neurotoxin heavy chains, light chains or variants thereof, modified botulinum neurotoxin chains and/or fragments of the chains. Cloned genes may also encode a targeting component.
  • the genes may be cloned into, for example, cloning vectors, such as phages or plasmids or phagemids.
  • cloning vectors such as phages or plasmids or phagemids.
  • the recombinant vectors are transformed into host cells, for example, into a prokaryotic cell, for example, E. coli . Proteins can be expressed and then isolated using conventional techniques.
  • Fusion genes may be used which encode more than one component of an agent.
  • a targeting component and a botulinum toxin heavy chain and/or light chain and/or a fragment of a heavy and/or a fragment of a light chain can be produced from a single cloned gene as a fusion protein.
  • individual components obtained from recombinant techniques can be chemically coupled to other components obtain from similar or other sources.
  • a targeting component may be coupled to a recombinant L chain or to a recombinant fusion LH N .
  • the linkages between the botulinum components and the targeting moieties may include appropriate spacer components, which may also be DNA encoded.
  • an LH N which may be a hybrid of an L chain and an H N from different botulinum toxin types, is expressed recombinantly as a fusion protein.
  • Such an LH N hybrid may also be coupled to a targeting component. There may be included one or more spacers between the L and H N and/or between the LH N and targeting component.
  • the L chain of a botulinum neurotoxin, or a fragment of the L chain containing the endopeptidase activity is expressed recombinantly to produce an agent for use in accordance with the present invention.
  • the L chain of a botulinum neurotoxin, or a fragment of the L chain containing the endopeptidase activity is expressed recombinantly as a fusion protein, with the H N of the H chain and the targeting component.
  • the expressed fusion protein may also include one or more spacer regions.
  • the L chain may be fused to H N which is in turn fused to the targeting component.
  • the H N may be fused to the L chain which is in turn fused to the targeting component.
  • Spacer components may be expressed recombinantly between some or all of the components of an agent of the invention.
  • the L chain is obtained from botulinum toxin type B and the amine end segment of the H N chain fragment is obtained from botulinum toxin type A.
  • the H N fragment of the botulinum toxin type A is produced according to the method described by Shone C. C., Hambleton, P., and Melling, J. (1987, Eur. J. Biochem. 167, 175-180) and the L chain of botulinum toxin type B according to the method of Sathyamoorthy, V. and DasGupta, B. R. (1985, J. Biol. Chem. 260, 10461-10466).
  • the free cysteine on the amine end segment of the H chain fragment of botulinum toxin type A is then derivatized by the addition of a ten-fold molar excess of dipyridyl disulphide followed by incubation at 4° C. overnight.
  • the excess dipvridyl disulphide and the thiopyridone by product are then removed by desalting the protein over a PD10 column (Pharmacia) into PBS.
  • the derivatized H N is then concentrated to a protein concentration in excess of 1 mg/ml before being mixed with an equimolar portion of L chain from botulinum toxin type B (>1 mg/ml in PBS). After overnight incubation at room temperature the mixture is separated by size exclusion chromatography over Superose 6 (Pharmacia), and the fractions analyzed by SDS-PAGE.
  • the chimeric LH N is then available to produce a conjugated agent which includes a targeting component.
  • the coupling of the targeting moieties to the botulinum components may be achieved via chemical coupling using reagents and techniques known to those skilled in the art.
  • any coupling chemistry capable of covalently attaching the targeting moieties of the agents to botulinum neurotoxin components and known to those skilled in the art is covered by the scope of this application.
  • a biological persistence enhancing component and/or a biological activity enhancing component may be added to a botulinum neurotoxin thereby increasing the biological persistence and/or biological activity of the botulinum neurotoxin.
  • a biological persistence enhancing component can be removed from a botulinum neurotoxin thereby decreasing the biological persistence and/or biological activity of the neurotoxin.
  • the botulinum neurotoxin can be a hybrid neurotoxin.
  • the neurotoxin's targeting, translocation and therapeutic components may be derived from different botulinum toxin serotypes.
  • the polypeptide may comprise a first amino acid sequence region derived from the H C of a botulinum toxin type A, a second amino acid sequence region derived from the H N Of botulinum type B, and a third amino acid sequence region derived from the light chain of botulinum serotype E. This is merely an example and all other possible combinations are included within the scope of the present invention.
  • the neurotoxin's targeting, translocation and therapeutic components can be modified from the naturally occurring sequence from which they are derived.
  • the amino acid sequence region can have at least one or more amino acids added, deleted or substituted as compared to the naturally occurring sequence.
  • Amino acids that can be substituted for amino acids contained in a biological persistence enhancing component include alanine, aspargine, cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, proline, glutamine, arginine, serine, threonine, valine, tryptophan, tyrosine and other naturally occurring amino acids as well as non-standard amino acids.
  • Methods of the present invention provide for reduction in restenosis ranging from about 1% to about 100% in effectiveness.
  • the reduction may be between about 1% and about 10% or between about 10% and about 20% or between about 10% and about 30% or between about 10% and about 40% or between about 10% and about 50% or between about 10% and about 60% or between about 10% and about 70% or between about 10% and about 80% or between about 10% and about 90% or between about 10% and about 100%.
  • the dose of neurotoxin to be administered may vary with the age, presenting condition and weight of the patient to be treated.
  • the potency of the neurotoxin will also be considered.
  • Toxin potency is expressed as a multiple of the LD 50 value for a mouse.
  • One “unit” of toxin can be defined as the amount of toxin that kills 50% of a group of mice that were disease-free prior to inoculation with the toxin.
  • commercially available Botulinum toxin A typically has a potency such that one nanogram contains about 40 mouse units.
  • the potency, or LD 50 in humans of the Botulinum toxin A product supplied by Allergan, Inc. under the registered trademark “BOTOX” is believed to be about 2,730 mouse units.
  • the neurotoxin can be administered in a dose of about 0.001 units up to about 100 units. In one embodiment, individual dosages of about 0.01 units to about 5 units are used. In another embodiment, individual dosages of about 0.01 units to about 3 units are used. In still another embodiment, individual dosages of about 0.01 units to about 1 unit are used. In still another embodiment, individual dosages of about 0.05 units to about 1 unit are used. Those of ordinary skill in the art will know, or can readily ascertain, how to adjust dosages for neurotoxin of greater or lesser potency in a certain circumstance.
  • potency may be expressed as a multiple of the LD 50 value of an agent of the invention for a mouse.
  • a “U” or “unit” of an agent can be defined as the amount of toxin that kills 50% of a group of mice that were disease-free prior to inoculation with the agent.
  • potency may be expressed as the LD 50 value of an agent that would be produced by an equal molar amount of a native, non-variant botulinum toxin.
  • the lowest therapeutically effective dosage will be administered to the patient.
  • the lowest therapeutic dosage is that dosage which results in the desired effect on a blood vessel of the patient to which the toxin is administered.
  • Methods for assessing or quantifying the effect of a toxin on a blood vessel can be determined by those skilled in the art. For example, use of optical coherence topography can provide a measure of the dilating effect of the toxin.
  • a low dosage may be administered to determine the patient's sensitivity to, and tolerance of, the neurotoxin. Additional administrations of the same or different dosages may be administered to the blood vessel as necessary.
  • a toxin may be administered to a blood vessel before a procedure, for example, a coronary angioplasty procedure, is performed, and/or during the procedure and/or after the procedure. The number of administrations and timing of the administrations may be determined by the treating physician.
  • the neurotoxins may be administered by, for example, injection into a blood vessel using a needle or by needleless injection.
  • the toxin may also be administered by application of the toxin to the wall of the blood vessel in a salve, lotion, ointment, cream, emulsion or the like delivery substrates.
  • an agent of the invention is administered to the patient by injection.
  • an agent may be injected into the cardiovascular system of a patient.
  • the injecting may be into an artery, for example, a coronary artery.
  • the injecting is into the region of the blood vessel where the vessel is to undergo a procedure, for example, a procedure to mechanically expand the internal diameter of an occluded blood vessel.
  • An agent may be injected into the wall of a blood vessel from outside of the blood vessel or an agent may be injected into the wall of a blood vessel from inside the blood vessel.
  • Methods for injecting into a wall of a blood vessel are well known to those of ordinary skill in the art.
  • an agent may be injected into the wall of a blood vessel from inside the blood vessel by the use of a catheter containing one or more needles for injecting.
  • microprojectile drug particles may be coated with a neurotoxin and then discharged into the blood vessel from an external delivery device. Depending on the discharge velocity and the distance from the injection site, the drug particles penetrate through the different layers of the blood vessels. As the microprojectiles penetrate through, or are deposited in, the blood vessel cells, the neurotoxin is released. Individual layers of blood vessels may be targeted for the microprojectiles.
  • the neurotoxins may also be administered using a stent or an angioplasty balloon that is coated or impregnated with the toxin, for example botulinum toxin.
  • U.S. Pat. Nos. 6,306,423 and 6,312,708 disclose material that may be impregnated, attached or imbedded with the toxin and which may be used to coat stents and/or angioplasty balloons. In addition, the material may be used to form stents which include the toxin. The disclosure of each of these two patents is incorporated in its entirety herein by reference.
  • the blood vessel to be treated is first administered with botulinum toxin before inserting the toxin comprising stent and/or balloon. In another embodiment, the blood vessel to be treated is not administered with toxin before inserting the botulinum toxin comprising stent and/or balloon.
  • botulinum toxin A administered into a blood vessel may produce a dilating and/or anti-inflammatory effect for, for example about 1 month to about 3 months, or for example, about 3 to about 6 months or for example from about 6 months to about 1 year.
  • the agent may be allowed to induce its effect on a blood vessel before a procedure, for example, a coronary angioplasty procedure, is performed.
  • the agent may be allowed to dilate the blood vessel and/or prevent inflammation of the blood vessel before a procedure is begun.
  • a physician of ordinary skill can determine when the agent has exerted its effect(s) on a blood vessel.
  • a 54 year old male patient complains of chest pains in an emergency room examination.
  • the patient smokes 2 to 3 packs of cigarettes a day, is of average weight and has a family history of coronary arterial blockage.
  • the patient is diagnosed as having an occlusion of the left coronary artery.
  • a coronary angiogram is used to measure the narrowing of the arteries. It is estimated that the patient suffers from an 80% occlusion of the left coronary artery. He is scheduled for a balloon angioplasty procedure for the following week.
  • the physician begins the procedure by injecting between about 0.05 units and about 5 units of botulinum toxin type A into the wall of the left coronary artery of the patient. Following injection, the artery is allowed to dilate. A 3-millimeter noncompliant balloon catheter is then inserted into the femoral artery of the patient through the groin/upper thigh area. The catheter is then fed through the artery up into the heart using a video monitor to guide the process. A guide wire is advanced to the location of the blocked artery, and the balloon catheter is passed along the guide wire into the target area of coronary blockage. When the catheter reaches the target area, the balloon is inflated for a period of several seconds to several minutes. After deflation of the balloon, the same area may be treated with one or more additional inflations. Examination reveals little or no damage to the treated artery.
  • a 62 year old male patient who is approximately 30% overweight and has a serum cholesterol level of approximately 260 complains of chest pains.
  • the patient is diagnosed as having coronary artery blockage and is scheduled for a percutaneous transluminal coronary angioplasty procedure.
  • botulinum toxin is injected directly into the wall of the artery in the area of the blockage. Following injection, the artery is allowed to dilate. A 3-millimeter compliant balloon catheter and stent are then inserted into the interosseous artery of the patient through the wrist area. The catheter and stent are then fed through the interosseous artery to the area of blockage. A guide wire is advanced to the location of the blocked artery, and the catheter and stent are passed along the guide wire into the target area of coronary blockage. When the catheter reaches the target area, the balloon is inflated and the stent is correspondingly expanded bracing open the artery. The balloon is deflated and removed leaving in place the expanded stent. There is no sign of damage to the artery.
  • a 49 year old male patient is diagnosed with coronary arterial blockage as a result of restenosis.
  • the patient has a history of coronary arterial blockage and has previously undergone a balloon angioplasty procedure. Six months after the procedure, the patient is diagnosed with an advanced case of restenosis.
  • the patient undergoes a percutaneous transluminal coronary angioplasty procedure in which botulinum toxin type A, B, C, D, D, F and/or G is used. Between about 0.1 units and about 4 units of botulinum toxin is injected into the wall of the blood vessel in the area of restenosis. Following injection, the artery is allowed to dilate. A 4-millimeter compliant balloon catheter and stent are then inserted into the femoral artery of the patient. The catheter and stent are then fed through the artery to the area of blockage using a video monitor to guide the process. A guide wire is advanced to the location of the blocked artery, and the catheter and stent are passed along the guide wire into the target area of coronary blockage.
  • the balloon When the catheter and stent reach the target area, the balloon is inflated and the stent correspondingly expanded holding open the artery. The balloon is deflated and removed leaving in place the expanded stent. 3 to 6 months after the procedure, the blood vessel is re-injected with the botulinum toxin in the area of the stent.
  • a 58 year old female patient is diagnosed with coronary arterial blockage.
  • the patient is scheduled for a percutaneous transluminal coronary angioplasty procedure in which a stent impregnated with botulinum toxin type A, B, C, D, E, F and/or G is used.
  • a botulinum toxin is injected into the wall of the blood vessel in the area of blockage. Following injection, the artery is allowed to dilate.
  • a 2-millimeter compliant balloon catheter and stent which is coated or impregnated with a botulinum toxin, are then inserted into the femoral artery of the patient.
  • the catheter and stent are passed through the femoral artery to the area of blockage using a video monitor to guide the process.
  • a guide wire is advanced to the location of the blocked artery, and the catheter and stent are passed along the guide wire into the target area of coronary blockage.
  • the balloon is inflated and the stent is correspondingly expanded bracing open the artery.
  • the balloon is deflated and removed leaving in place the expanded stent.
  • a 50 year old male patient is diagnosed with coronary arterial blockage of the left coronary artery and is scheduled for a percutaneous transluminal coronary angioplasty procedure in which a self expanding stent impregnated with a botulinum toxin is used.
  • the physician begins the procedure by injecting between about 0.1 units and about 5 units of botulinum toxin type A into the wall of the left coronary artery of the patient. Following injection, the artery is allowed to dilate. A self expanding stent impregnated with the botulinum toxin is then inserted with a catheter into the common interosseous artery of the patient through the wrist area. The catheter and stent are passed through the interosseous artery to the area of blockage. A guide wire is advanced to the location of the blocked artery advancing the botulinum toxin impregnated, self expanding stent into the target area of coronary blockage. When the catheter reaches the target area, the stent is expanded bracing open the artery.
  • a 51 year old female patient complains of chest pains.
  • the patient is overweight and has a serum cholesterol level of approximately 270.
  • the patient is diagnosed as having coronary artery blockage.
  • a coronary angiogram is used to measure the narrowing of the arteries. It is estimated that the patient suffers from a 70% to 90% occlusion of a coronary artery. She is scheduled for a percutaneous transluminal coronary angioplasty procedure.
  • a botulinum toxin is injected directly into the wall of the artery in the area of the blockage.
  • a catheter which includes one or more injection needles is inserted through the femoral artery of the patient through the groin/upper thigh area into the area of the coronary blockage. There the botulinum toxin is injected into the inner wall of the occluded blood vessel.
  • the artery is allowed to dilate.
  • a 3-millimeter compliant balloon catheter and stent impregnated with botulinum toxin type A are then inserted into the femoral artery of the patient.
  • the catheter and stent are fed through the femoral artery to the area of blockage using a video monitor to guide the process.
  • a guide wire is advanced to the location of the blocked artery, and the catheter and stent is passed along the guide wire into the target area of coronary blockage.
  • the balloon is inflated and the stent is correspondingly expanded bracing open the artery.
  • the balloon is deflated and removed leaving in place the expanded stent. There is no sign of damage to the blood vessel.
  • My invention also includes within its scope the use of a neurotoxin, such as a botulinum toxin, in the preparation of a medicament for the treatment of a cardiovascular disease.
  • a neurotoxin such as a botulinum toxin

Abstract

The present invention provides for methods of treating cardiovascular diseases in a mammal. The methods include a step of administering an effective amount of a botulinum toxin directly to a blood vessel of a mammal thereby treating a cardiovascular disease.

Description

CROSS REFERENCE
This application is a continuation in part of application Ser. No. 09/371,352, filed Aug. 10, 1999, now U.S. Pat. No. 6,263,040 the entire content of which prior patent application is incorporated herein by reference in its' entity.
BACKGROUND
The present invention relates to methods of preventing or reducing restenosis that may occur in blood vessels after mechanically expanding the diameter of an occluded blood vessel. Atherosclerosis is a progressive disease wherein fatty, fibrous, calcific, or thrombotic deposits produce atheromatous plaques, within and beneath the intima which is the innermost layer of arteries. Atherosclerosis tends to involve large and medium sized arteries. The most commonly affected are the aorta, iliac, femoral, coronary, and cerebral arteries. Clinical symptoms occur because the mass of the atherosclerotic plaque reduces blood flow through the afflicted artery, thereby compromising tissue or organ function distal to it.
Percutaneous transluminal coronary angioplasty is a non-surgical method for treatment of coronary atherosclerosis. In this procedure, an inflatable balloon is inserted in a coronary artery in the region of arterial narrowing. Inflation of the balloon for 15-30 seconds results in an expansion of the narrowed lumen or passageway. Because residual narrowing is usually present after the first balloon inflation, multiple or prolonged inflations are routinely performed to reduce the severity of the residual tube narrowing.
Stents are often used in combination with coronary balloon angioplasty. Typically, a stent is used to brace the blood vessel open after an initial expansion of the narrowed blood vessel by a balloon. Self expanding stents are also used to expand and hold open occluded blood vessels. Various stents and their use are disclosed in U.S. Pat. Nos. 6,190,404; 6,344,055; 6,306,162; 6,293,959; 6,270,521; 6,264,671; 6,261,318; 6,241,758; 6,217,608; 6,196,230; 6,183,506; 5,989,280. The disclosure of each of these patents is incorporated in its entirety herein by reference.
One problem with angioplasty is that following the procedure restenosis, or recurrence of the obstruction, may occur. Tears in the wall expose blood to foreign material and proteins, such as collagen, which are highly thrombogenic. Resulting clots can contain growth hormones which may be released by platelets within the clot. Additionally, thrombosis may cause release of growth hormones and cytokines by cells from macrophages. Growth hormones may cause smooth muscle cells and fibroblasts to aggregate in the region and multiply. Further, following angioplasty there is often a loss of the single layer of cells that normally covers the internal surface of blood vessels which leads to thrombosis. The combination of tearing of the blood vessel wall and the loss of the endothelial layer often generates an internal blood vessel surface which is quite thrombogenic. Restenosis may result from the proliferation of smooth muscle cells, which normally reside within the arterial wall, in the area of the injury in response to the thrombosis.
Angioplasty procedures also produce injuries in the arterial wall which become associated with inflammation. Any kind of inflammatory response may cause growth of new tissue, for example, scar tissue, which may contribute to restenosis.
One of the other major causes of restenosis following angioplasty may be that the injured arterial wall may exhibit a reduced hemocompatability compared to that associated with a normal arterial wall. Adverse responses which are associated with reduced hemocompatability include platelet adhesion, aggregation, and activation; thrombosis; inflammatory cell reactions such as adhesion and activation of monocytes or macrophages; and the infiltration of leukocytes into the arterial wall.
Restenosis is a serious problem that may occur in over one third of all coronary angioplasty patients. Therefore, there exists a need for methods to reduce or eliminate the occurrence of restenosis which may follow procedures to mechanically expand an occluded blood vessel.
Botulinum Toxin
The anaerobic, Gram positive bacterium Clostridium botulinum produces a potent polypeptide neurotoxin, botulinum toxin, which causes a neuroparalytic illness in humans and animals referred to as botulism. The spores of Clostridium botulinum are found in soil and can grow in improperly sterilized and sealed food containers of home based canneries, which are the cause of many of the cases of botulism. The effects of botulism typically appear 18 to 36 hours after eating the foodstuffs infected with a Clostridium botulinum culture or spores. The botulinum toxin can apparently pass unattenuated through the lining of the gut and attack peripheral motor neurons. Symptoms of botulinum toxin intoxication can progress from difficulty walking, swallowing, and speaking to paralysis of the respiratory muscles and death.
Botulinum toxin type A (“BoNT/A”) is the most lethal natural biological agent known to man. About 50 picograms of botulinum toxin (purified neurotoxin complex) serotype A is a LD50 in mice. One unit (U) of botulinum toxin is defined as the LD50 upon intraperitoneal injection into female Swiss Webster mice weighing 18-20 grams each. Seven immunologically distinct botulinum neurotoxins have been characterized, these being respectively botulinum neurotoxin serotypes A, B, C1, D, E, F and G each of which is distinguished by neutralization with serotype-specific antibodies. The different serotypes of botulinum toxin vary in the animal species that they affect and in the severity and duration of the paralysis they evoke. For example, it has been determined that BoNt/A is 500 times more potent, as measured by the rate of paralysis produced in the rat, than is botulinum toxin serotype B (BoNT/B). Additionally, botulinum toxin type B (“BoNt/B”) has been determined to be non-toxic in primates at a dose of 480 U/kg which is about 12 times the primate LD50 for BoNt/A. Botulinum toxin apparently binds with high affinity to cholinergic motor neurons, is translocated into the neuron and blocks the release of acetylcholine.
Botulinum toxins have been used in clinical settings for the treatment of neuromuscular disorders characterized by hyperactive skeletal muscles. BoNt/A has been approved by the U.S. Food and Drug Administration for the treatment of blepharospasm, strabismus, hemifacial spasm and cervical dystonia. Additionally, a botulinum toxin type B has been approved by the FDA for the treatment of cervical dystonia. Non-serotype A botulinum toxin serotypes apparently have a lower potency and/or a shorter duration of activity as compared to BoNt/A. Clinical effects of peripheral intramuscular BoNt/A are usually seen within one week of injection. The typical duration of symptomatic relief from a single intramuscular injection of BoNt/A averages about three months.
Although all the botulinum toxins serotypes apparently inhibit release of the neurotransmitter acetylcholine at the neuromuscular junction, they do so by affecting different neurosecretory proteins and/or cleaving these proteins at different sites. For example, botulinum serotypes A and E both cleave the 25 kiloDalton (kD) synaptosomal associated protein (SNAP-25), but they target different amino acid sequences within this protein. BoNT/B, D, F and G act on vesicle-associated protein (VAMP, also called synaptobrevin), with each serotype cleaving the protein at a different site. Finally, botulinum toxin serotype C1 (BoNT/C1) has been shown to cleave both syntaxin and SNAP-25. These differences in mechanism of action may affect the relative potency and/or duration of action of the various botulinum toxin serotypes.
Regardless of serotype, the molecular mechanism of toxin intoxication appears to be similar and to involve at least three steps or stages. In the first step of the process, the toxin binds to the presynaptic membrane of the target neuron through a specific interaction between the H chain and a cell surface receptor; the receptor is thought to be different for each serotype of botulinum toxin and for tetanus toxin. The carboxyl end segment of the H chain, HC, appears to be important for targeting of the toxin to the cell surface.
In the second step, the toxin crosses the plasma membrane of the poisoned cell. The toxin is first engulfed by the cell through receptor-mediated endocytosis, and an endosome containing the toxin is formed. The toxin then escapes the endosome into the cytoplasm of the cell. This last step is thought to be mediated by the amino end segment of the H chain, HN, which triggers a conformational change of the toxin in response to a pH of about 5.5 or lower. Endosomes are known to possess a proton pump which decreases intra endosomal pH. The conformational shift exposes hydrophobic residues in the toxin, which permits the toxin to embed itself in the endosomal membrane. The toxin then translocates through the endosomal membrane into the cytosol.
The last step of the mechanism of botulinum toxin activity appears to involve reduction of the disulfide bond joining the H and L chain. The entire toxic activity of botulinum and tetanus toxins is contained in the L chain of the holotoxin; the L chain is a zinc (Zn++) endopeptidase which selectively cleaves proteins essential for recognition and docking of neurotransmitter-containing vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane. Tetanus neurotoxin, botulinum toxin/B/D,/F, and/G cause degradation of synaptobrevin (also called vesicle-associated membrane protein (VAMP)), a synaptosomal membrane protein. Most of the VAMP present at the cytosolic surface of the synaptic vesicle is removed as a result of any one of these cleavage events. Each toxin specifically cleaves a different bond.
The molecular weight of the botulinum toxin protein molecule, for all seven of the known botulinum toxin serotypes, is about 150 kD. Interestingly, the botulinum toxins are released by Clostridial bacterium as complexes comprising the 150 kD botulinum toxin protein molecule along with associated non-toxin proteins. Thus, the BoNt/A complex can be produced by Clostridial bacterium as 900 kD, 500 kD and 300 kD forms. BoNT/B and C1 are apparently produced as only a 500 kD complex. BoNT/D is produced as both 300 kD and 500 kD complexes. Finally, BoNT/E and F are produced as only approximately 300 kD complexes. The complexes (i.e. molecular weight greater than about 150 kD) are believed to contain a non-toxin hemaglutinin protein and a non-toxin and non-toxic nonhemaglutinin protein. These two non-toxin proteins (which along with the botulinum toxin molecule comprise the relevant neurotoxin complex) may act to provide stability against denaturation to the botulinum toxin molecule and protection against digestive acids when toxin is ingested. Additionally, it is possible that the larger (greater than about 150 kD molecular weight) botulinum toxin complexes may result in a slower rate of diffusion of the botulinum toxin away from a site of intramuscular injection of a botulinum toxin complex.
In vitro studies have indicated that botulinum toxin inhibits potassium cation induced release of both acetylcholine and norepinephrine from primary cell cultures of brainstem tissue. Additionally, it has been reported that botulinum toxin inhibits the evoked release of both glycine and glutamate in primary cultures of spinal cord neurons and that in brain synaptosome preparations botulinum toxin inhibits the release of each of the neurotransmitters acetylcholine, dopamine, norepinephrine, CGRP and glutamate.
BoNt/A can be obtained by establishing and growing cultures of Clostridium botulinum in a fermenter and then harvesting and purifying the fermented mixture in accordance with known procedures. All the botulinum toxin serotypes are initially synthesized as inactive single chain proteins which must be cleaved or nicked by proteases to become neuroactive. The bacterial strains that make botulinum toxin serotypes A and G possess endogenous proteases and serotypes A and G can therefore be recovered from bacterial cultures in predominantly their active form. In contrast, botulinum toxin serotypes C1, D and E are synthesized by nonproteolytic strains and are therefore typically unactivated when recovered from culture. Serotypes B and F are produced by both proteolytic and nonproteolytic strains and therefore can be recovered in either the active or inactive form. However, even the proteolytic strains that produce, for example, the BoNt/B serotype only cleave a portion of the toxin produced. The exact proportion of nicked to unnicked molecules depends on the length of incubation and the temperature of the culture. Therefore, a certain percentage of any preparation of, for example, the BoNt/B toxin is likely to be inactive, possibly accounting for the known significantly lower potency of BoNt/B as compared to BoNt/A. The presence of inactive botulinum toxin molecules in a clinical preparation will contribute to the overall protein load of the preparation, which has been linked to increased antigenicity, without contributing to its clinical efficacy. Additionally, it is known that BoNt/B has, upon intramuscular injection, a shorter duration of activity and is also less potent than BoNt/A at the same dose level.
It has been reported (as exemplary examples) that BoNt/A has been used clinically as follows:
(1) about 75-125 units of BOTOX®1 per intramuscular injection (multiple muscles) to treat cervical dystonia;
1 Available from Allergan, Inc., of Irvine, Calif. under the tradename BOTOX®.
(2) 5-10 units of BOTOX® per intramuscular injection to treat glabellar lines (brow furrows) (5 units injected intramuscularly into the procerus muscle and 10 units injected intramuscularly into each corrugator supercilii muscle);
(3) about 30-80 units of BOTOX® to treat constipation by intrasphincter injection of the puborectalis muscle;
(4) about 1-5 units per muscle of intramuscularly injected BOTOX® to treat blepharospasm by injecting the lateral pre-tarsal orbicularis oculi muscle of the upper lid and the lateral pre-tarsal orbicularis oculi of the lower lid.
(5) to treat strabismus, extraocular muscles have been injected intramuscularly with between about 1-5 units of BOTOX®, the amount injected varying based upon both the size of the muscle to be injected and the extent of muscle paralysis desired (i.e. amount of diopter correction desired).
(6) to treat upper limb spasticity following stroke by intramuscular injections of BOTOX® into five different upper limb flexor muscles, as follows:
(a) flexor digitorum profundus: 7.5 U to 30 U
(b) flexor digitorum sublimus: 7.5 U to 30 U
(c) flexor carpi ulnaris: 10 U to 40 U
(d) flexor carpi radialis: 15 U to 60 U
(e) biceps brachii: 50 U to 200 U. Each of the five indicated muscles has been injected at the same treatment session, so that the patient receives from 90 U to 360 U of upper limb flexor muscle BOTOX® by intramuscular injection at each treatment session.
The tetanus neurotoxin acts mainly in the central nervous system, while botulinum neurotoxin acts at the neuromuscular junction; both act by inhibiting acetylcholine release from the axon of the affected neuron into the synapse, resulting in paralysis. The effect of intoxication on the affected neuron is long-lasting and until recently has been thought to be irreversible. The tetanus neurotoxin is known to exist in one immunologically distinct serotype.
Acetylcholine
Typically only a single type of small molecule neurotransmitter is released by each type of neuron in the mammalian nervous system. The neurotransmitter acetylcholine is secreted by neurons in many areas of the brain, but specifically by the large pyramidal cells of the motor cortex, by several different neurons in the basal ganglia, by the motor neurons that innervate the skeletal muscles, by the preganglionic neurons of the autonomic nervous system (both sympathetic and parasympathetic), by the postganglionic neurons of the parasympathetic nervous system, and by some of the postganglionic neurons of the sympathetic nervous system. Essentially, only the postganglionic sympathetic nerve fibers to the sweat glands, the piloerector muscles and a few blood vessels are cholinergic and most of the postganglionic neurons of the sympathetic nervous system secret the neurotransmitter norepinephine. In most instances acetylcholine has an excitatory effect. However, acetylcholine is known to have inhibitory effects at some of the peripheral parasympathetic nerve endings, such as inhibition of the heart by the vagal nerve.
The efferent signals of the autonomic nervous system are transmitted to the body through either the sympathetic nervous system or the parasympathetic nervous system. The preganglionic neurons of the sympathetic nervous system extend from preganglionic sympathetic neuron cell bodies located in the intermediolateral horn of the spinal cord. The preganglionic sympathetic nerve fibers, extending from the cell body, synapse with postganglionic neurons located in either a paravertebral sympathetic ganglion or in a prevertebral ganglion. Since, the preganglionic neurons of both the sympathetic and parasympathetic nervous system are cholinergic, application of acetylcholine to the ganglia will excite both sympathetic and parasympathetic postganglionic neurons.
Acetylcholine activates two types of receptors, muscarinic and nicotinic receptors. The muscarinic receptors are found in all effector cells stimulated by the postganglionic neurons of the parasympathetic nervous system, as well as in those stimulated by the postganglionic cholinergic neurons of the sympathetic nervous system. The nicotinic receptors are found in the synapses between the preganglionic and postganglionic neurons of both the sympathetic and parasympathetic. The nicotinic receptors are also present in many membranes of skeletal muscle fibers at the neuromuscular junction.
Acetylcholine is released from cholinergic neurons when small, clear, intracellular vesicles fuse with the presynaptic neuronal cell membrane. A wide variety of non-neuronal secretory cells, such as, adrenal medulla (as well as the PC12 cell line) and pancreatic islet cells release catecholamines and insulin, respectively, from large dense-core vesicles. The PC12 cell line is a clone of rat pheochromocytoma cells extensively used as a tissue culture model for studies of sympathoadrenal development. Botulinum toxin inhibits the release of both types of compounds from both types of cells in vitro, permeabilized (as by electroporation) or by direct injection of the toxin into the denervated cell. Botulinum toxin is also known to block release of the neurotransmitter glutamate from cortical synaptosomes cell cultures.
SUMMARY
The present invention provides for methods to treat cardiovascular diseases in a mammal, for example, in a human.
The methods include a step of administering an effective amount of a botulinum toxin directly to a blood vessel of a mammal to treat a cardiovascular disease. In one embodiment, treating the cardiovascular disease prevents restenosis.
In one embodiment of the invention, the mammal is having or has had a cardiovascular procedure. In one embodiment, the cardiovascular procedure is an arterial cardiovascular procedure, for example, a coronary arterial cardiovascular procedure.
In one embodiment, the cardiovascular procedure includes an angioplasty procedure. In one embodiment, the angioplasty includes the step of inserting a stent into the blood vessel of the mammal. In another embodiment, the angioplasty does not include the step of inserting a stent into a blood vessel. The angioplasty procedure may be, for example, balloon angioplasty. In one embodiment, the balloon angioplasty includes the use of a stent. For example, a stent may be inserted into the blood vessel during the balloon angioplasty.
The procedure is not limited to use of a balloon. Any device that may be used to mechanically open a constricted blood vessel, for example, a spring or other expanding device, may be used to perform an angioplasty.
The step of administering the botulinum toxin may include a step of injecting the botulinum toxin into a wall of the blood vessel. In particular, the toxin may be injected into the intima, media and/or adventia layers of the blood vessel. Further, the step of administering may be accomplished using a stent which has been coated or impregnated with botulinum toxin.
In one embodiment of the present invention, the botulinum toxin reduces or eliminates damage to a blood vessel. Examples of damage that may occur are stretching and/or tearing of a blood vessel or any other damage that may occur to the blood vessel as a result of mechanically expanding the inner diameter of the blood vessel. In one embodiment, the botulinum toxin reduces or eliminates damage to the blood vessel, at least in part, by dilating the blood vessel. In another embodiment, the botulinum toxin reduces or eliminates damage to the blood vessel, at least in part, by reducing or eliminating inflammation of the blood vessel.
In accordance with the present invention, the botulinum toxin may be any botulinum toxin including botulinum toxin types A, B, C, D, E, F, G or mixtures thereof or combinations thereof, including a modified, hybrid or chimeric botulinum toxin.
Further in accordance with the present invention, there are provided methods to prevent restenosis in a blood vessel in a mammal which may occur following a cardiovascular procedure. In one embodiment, the method includes a step of administering to the mammal an effective amount of botulinum toxin thereby preventing restenosis in a blood vessel.
Still further in accordance with the present invention there are provided methods to prevent restenosis in a mammal by preventing damage in a blood vessel which may occur during or following a cardiovascular procedure. In one embodiment, the method includes the step of administering to a mammal an effective amount of a botulinum toxin thereby preventing damage in the blood vessel and preventing restenosis.
The present invention also provides for methods to prevent restenosis in a mammal by preventing inflammation in a blood vessel which may occur during or following a cardiovascular procedure. In one embodiment, the method includes the step of administering to a mammal an effective amount of the botulinum toxin thereby preventing inflammation in the blood vessel and preventing restenosis.
Further, the present invention provides for methods to prevent restenosis in a mammal by dilating a blood vessel proceeding, during or following a cardiovascular procedure. In one embodiment, the method includes the step of administering to a mammal an effective amount of the botulinum toxin thereby dilating the blood vessel and preventing restenosis.
Still further, the present invention provides for compositions for use in cardiovascular procedures. In one embodiment, these compositions include a stent with a botulinum toxin attached to the stent or imbedded in the stent. The botulinum toxin may be any botulinum toxin including botulinum toxin type A, B, C, D, E, F, G or combinations thereof or mixtures thereof.
Any feature or combination of features described herein are included within the scope of the present invention provided that the features included in any such combination are not mutually inconsistent as will be apparent from the context, this specification, and the knowledge of one of ordinary skill in the art.
Additional advantages and aspects of the present invention are apparent in the following detailed description and claims.
DEFINITIONS
“Agent” is defined as a neurotoxin, for example, a botulinum toxin, for use in accordance with the present invention. An agent may be a fragment of a neurotoxin, a modified neurotoxin or a variant neurotoxin that possesses some or all of the biological activity of an unmodified neurotoxin.
“Angioplasty” means any procedure where the inner diameter of a blood vessel is mechanically expanded.
A “botulinum toxin” may refer to native botulinum toxin or a functional fragment of a botulinum toxin or a modified botulinum toxin. In addition, botulinum toxins with amino acid deletions, additions, alterations or substitutions that delete, add, alter or substitute a single amino acid, or a small percentage of amino acids (for example, less than about 5%, or for example, less than about 1%) are conservatively modified variations of botulinum toxins. Where one or more substitutions of an amino acid(s) with a chemically similar amino acid are made in a botulinum toxin, this also results in a conservatively modified variation of a botulinum toxin. Tables providing functionally similar amino acids are well known in the art. The following five groups each contain amino acids that are conservative substitutions for one another: Aliphatic: Glycine (G), Alanine (A), Valine (V), Leucine (L), Isoleucine (I); Aromatic: Phenylalanine (F), Tyrosine (Y), Tryptophan (W); Sulfur-containing: Methionine (M), Cysteine (C); Basic: Arginine (R), Lysine (K), Histidine (H); Acidic: Aspartic acid (D), Glutamic acid (E), Asparagine (N), Glutamine (Q). See also, Creighton (1984) Proteins, W. H. Freeman and Company. Conservatively modified variations of native botulinum toxins are included within the scope of the meaning of “botulinum toxin.”
“Cardiovascular” means pertaining to blood vessels, for example, blood vessels of the heart.
“Clostridial toxin” or “Clostridial neurotoxin” means a toxin produced naturally by the genus of bacteria Clostridium. For example, Clostidial toxins include, but are not limited to, botulinum toxins, tetanus toxins, difficile toxins and butyricum toxins. A Clostridial toxin can also be made by known recombinant means by a non-Clostridial bacterium.
“Combination” means an ordered sequence of elements. For example, a combination of botulinum toxins may mean administration of botulinum toxin E, followed by administration of botulinum toxin type A, followed by administration of botulinum toxin type B. This is opposed to a “mixture” where, for example, different toxin types are combined prior to administration.
“Damage” means tearing, scratching, stretching, scraping, bruising and/or inflammation or injury caused by inflammation or other injury that may occur in a blood vessel undergoing a procedure, for example, a procedure where the inner diameter of the blood vessel is expanded using mechanical force.
“Fragment” means an amino acid sequence that comprises five amino acids or more of the native amino acid sequence up to a size of minus at least one amino acid from the native sequence. For example, a fragment of a botulinum toxin type A light chain comprises five or more amino acids of the amino acid sequence of the native botulinum toxin type A light chain up to a size of minus one amino acid from the native light chain. “HC” means a fragment obtained from the H chain of a Clostridial toxin which is equivalent, for example functionally equivalent, to the carboxyl end fragment of the H chain, or the portion corresponding to that fragment in the intact H chain involved in binding to a cell surface or cell surface receptor.
“HN” means a fragment or variant obtained from an H chain of a Clostridial toxin which may be functionally equivalent to the portion of an intact H chain involved in the translocation of at least the L chain across an intracellular endosomal membrane into a cytoplasm of a cell. An HN, may result from an HC being removed from an H chain. An HN may also result from an H chain being modified such that its HC no longer binds to cholinergic cell surfaces.
“Heavy chain” means the heavy chain of a Clostridial neurotoxin or a fragment or variant of an HN of a Clostridial neurotoxin. A heavy chain may have a molecular weight of about 100 kD and can be referred to as H chain, or as H.
“LHN” means a fragment obtained from a Clostridial neurotoxin that contains the L chain coupled to an HN. LHN can be obtained from the intact Clostridial neurotoxin by proteolysis, so as to remove or to modify the HC domain.
“Light chain” means the light chain of a Clostridial neurotoxin or a fragment or variant of a light chain of a Clostridial neurotoxin. A light chain may have a molecular weight of about 50 kD, and can be referred to as L chain, L, or as the proteolytic domain of a Clostridial neurotoxin.
“Linker” means a molecule which couples two or more other molecules or components together.
A “modified neurotoxin” means a neurotoxin that has a nonnative component covalently attached to the neurotoxin and/or a native portion of the neurotoxin missing. For example, a modified botulinum toxin may be a light chain of a botulinum toxin with a substance P molecule covalently attached.
“Neurotoxin” or “toxin” means a substance that inhibits neuronal function or cellular secretion. Clostridial toxins are examples of a neurotoxin.
“Prevent” means to keep from occurring in whole or in part.
“Reduce” means to make smaller in magnitude (e.g. size, quantity or number). The reduction may be about 1% to about 100%. For example, the reduction may be between about 1% and about 10% or between about 10% and about 20% or between about 10% and about 30% or between about 10% and about 40% or between about 10% and about 50% or between about 10% and about 60% or between about 10% and about 70% or between about 10% and about 80% or between about 10% and about 90% or between about 10% and about 100%.
“Spacer” means a molecule or set of molecules which physically separate and/or add distance between components of agents for use in accordance with the invention.
“Substantially” means largely but not entirely. For example, substantially may mean about 10% to about 99.999%, about 20% to about 99.999%, about 30% to about 99.999%, about 40% to about 99.999% or about 50% to about 99.999%.
“Targeting component” means a molecule that has a specific binding affinity for a cell surface or cell surface receptor.
“Variant” means a molecule or peptide which is substantially the same as that of a disclosed molecule or peptide in its structure and function. For example, a variant of a specified light chain may have amino acid sequence differences when compared to the amino acid sequence of the specified light chain. Variants may be considered to be equivalent to the specifically disclosed molecules and as such are within the scope of the invention.
DESCRIPTION
The present invention is, in part, based upon the discovery that a neurotoxin, for example, botulinum toxin, is useful for treating cardiovascular disease, for example, treating cardiovascular disease in a patient who has undergone, or is undergoing, a cardiovascular procedure. In one embodiment, the present invention provides for methods to reduce or eliminate restenosis following a cardiovascular procedure.
One skilled in the art will appreciate that the herein disclosed methods may find application in any blood vessel in the body including, but not limited to, coronary (heart), cerebral (brain), Carotid (neck), Renal (kidney), Visceral (abdominal), Iliac (hip), Femoropopliteal (thigh), Infrapopliteal (knee) blood vessels.
The invention comprises application of a neurotoxin, for example, a botulinum toxin, to a blood vessel of a patient who is undergoing or will undergo or has undergone a procedure that may, directly or indirectly, lead to damage of a blood vessel, for example, a coronary artery. In one embodiment, the present invention provides for methods to treat a patient undergoing an angioplasty procedure such that restenosis is reduced or eliminated following the procedure. In one embodiment, the angioplasty includes use of a stent, for example, a self expanding stent. In another embodiment, the angioplasty is balloon angioplasty. In another embodiment, the angioplasty is balloon angioplasty that includes use of a stent. In another embodiment, the angioplasty is balloon angioplasty that does not include use of a stent.
Without wishing to limit the present invention to any theory or mechanism of operation, it is thought that the present methods prevent damage to a blood vessel which may occur in association with a mechanical expanding of an otherwise occluded or partially occluded blood vessel. Therefore, methods of the present invention may prevent restenosis that may otherwise occur as a result of such damage. Examples of damage that may be prevented are tearing, scratching, stretching, scraping, bruising and/or inflammation or injury caused by inflammation or other injury that may occur in a blood vessel undergoing a procedure, for example, a procedure where the inner diameter of the blood vessel is expanded using mechanical force.
Though the mechanism of operation of botulinum toxin in preventing damage from occurring in a blood vessel is not completely understood, without wishing to limit the invention to any particular theory or mechanism of operation, the inventor suggests at least two possible theories of operation.
In one instance, the toxins are thought exert a dilating effect on blood vessels thereby increasing the diameter of a vessel, including the inner diameter of the vessel. Optical coherence topography may be used to provide a measure of the dilating effect of the toxin. The dilating effect of the toxin may be quantitated as a factor of the original size of the blood vessel opening before administration of the toxin. In one embodiment, the blood vessel opening may be dilated to between about 1.5× and about 100× the size of the opening before administration of the toxin. For example, the blood vessel opening may be dilated to between about 2× and about 5× the size of the opening before administration of the toxin. In another example the blood vessel opening may be dilated to between about 2× and about 10× the size of the opening before administration of the toxin. In another example the blood vessel opening may be dilated to between about 2× and about 30× the size of the opening before administration of the toxin. In another example the blood vessel opening may be dilated to between about 2× and about 50× the size of the opening before administration of the toxin. In another example the blood vessel opening may be dilated to between about 50× and about 100× the size of the opening before administration of the toxin.
The dilating of the blood vessels may make the vessels more receptive to intervention procedures. For example, balloon angioplasty and/or insertion of a stent or other mechanical intervention may be less likely to damage the blood vessel when the blood vessel is in the dilated state. After administration of an agent to the blood vessel, the blood vessel may be allowed to dilate before the procedure, for example, an angioplasty procedure, is performed. Whether dilation has taken place, and to what extent dilation has taken place can be determined by a physician of ordinary skill. For example, optical coherence topography may be used to make these determinations.
In another non-limiting theory of operation, it is believed that the toxins disclosed herein act on inflammation mediating cells, for example, blood vessel endothelial cells. These cells present many biologically active inflammation mediators which may include bradykinin, nitric oxide and vasoactive intestinal peptide. Release of these and/or other mediators may contribute to the events which cause blood vessel inflammation which may contribute to restenosis.
During secretion or exocytosis, the mediators may be included in vesicles which fuse to the inner surface of the cell membrane thereby releasing the vesicle contents to the outside of the cell. It is theorized that interference with the exocytosis process may be the mode of action of the Clostridial toxins.
It is theorized that Clostridial toxins may operate by preventing or reducing the secretion of inflammation producing molecules in blood vessel cells or other cells by cleaving or by otherwise interfering with the function of proteins involved in the secretory process by use of a light chain component, for example, a botulinum light chain component. A heavy chain component, for example HN, may also function in certain embodiments of the present invention by, for example, assisting in the release of an agent of the invention from intracellular vesicles, for example, endosomes.
Without wishing to limit the invention to any theory or mechanism of operation, it is conjectured that inflammation may either directly, or indirectly contribute to restenosis. By preventing or reducing blood vessel inflammation that may be associated with cardiovascular procedures, for example, balloon angioplasty and/or insertion of a stent, restenosis may be reduced in a patient who has undergone a cardiovascular procedure.
Another possible mechanism for the efficacy of the present disclosed invention is an effect of a botulinum toxin to inhibit neuronally mediated blood vessel contraction. Pretreatment with a botulinum toxin can inhibit a post stretch constriction. Within the scope of the present invention is a botulinum toxin which is a targeted toxin wherein the native binding moiety of the toxin has been replaced in whole or in part by a new binding moiety which targets the toxin to alpha2 receptors on sympathetic neurons which innervate the blood vessel to be treated. Furthermore, NO can be induced locally to cause dilation.
The neurotoxin for use in accordance with the present invention may comprise a targeting component, a therapeutic component and a translocation component.
In one embodiment, the targeting component comprises a carboxyl end fragment of a heavy chain of a butyricum toxin, a tetani toxin or a botulinum toxin including botulinum toxin types A, B, C, D, E, F and G.
In another embodiment the targeting component may be of non-botulinum toxin origin. Examples of targeting components that may be used in the present invention include, but are not limited to, antibodies, monoclonal antibodies, antibody fragments (Fab, F(ab)′2, Fv, ScFv, and other antibody fragments of the like), lectins, hormones, cytokines, growth factors, peptides, carbohydrates, lipids, glycons and nucleic acids. Other targeting components that may be useful in accordance with the present invention are disclosed in WO 01/21213 which is incorporated in its entirety herein by reference.
One exemplary targeting component for use in accordance with the present invention is substance P or substances similar to substance P. Use of substance P, or substances similar to substance P, as targeting components is described in U.S. patent application Ser. Nos. 09/489,667; 09/922,093 and 09/625,098 each of which is incorporated in its entirety herein by reference.
The therapeutic component operates to selectively cleave proteins essential for recognition and docking of secretory vesicles with the cytoplasmic surface of the plasma membrane, and fusion of the vesicles with the plasma membrane. One effect of the therapeutic component may be to substantially interfere with the release of neurotransmitters from a cell. Another effect of the therapeutic component may be to cause dilation of blood vessels. Another effect may be to cause flaccid paralysis of smooth muscle tissue. Another effect may be to reduce or eliminate secretion from cells, for example, inflammation producing cells. In one embodiment, the therapeutic component comprises a light chain of a butyricum toxin, a tetani toxin, a botulinum toxin, for example, botulinum toxin type A, B, C, D, E, F and G.
The translocation component may facilitate the transfer of at least a part of the neurotoxin, for example the therapeutic component into the cytoplasm of the target cell. In one embodiment, the translocation component comprises an amino end fragment of a heavy chain of a butyricum toxin, a tetani toxin, a botulinum toxin, for example, botulinum toxin type A, B, C, D, E, F and G.
According to a broad aspect of this invention, recombinant DNA methodologies may be used to produce the components of agents useful in accordance with the invention. These techniques may include steps of obtaining cloned genes from natural sources, or from synthetic oligonucleotide sequences, which may encode botulinum neurotoxin components including botulinum neurotoxin heavy chains, light chains or variants thereof, modified botulinum neurotoxin chains and/or fragments of the chains. Cloned genes may also encode a targeting component.
The genes may be cloned into, for example, cloning vectors, such as phages or plasmids or phagemids. The recombinant vectors are transformed into host cells, for example, into a prokaryotic cell, for example, E. coli. Proteins can be expressed and then isolated using conventional techniques.
Fusion genes may be used which encode more than one component of an agent. For example, a targeting component and a botulinum toxin heavy chain and/or light chain and/or a fragment of a heavy and/or a fragment of a light chain, can be produced from a single cloned gene as a fusion protein. Alternatively, individual components obtained from recombinant techniques can be chemically coupled to other components obtain from similar or other sources. For example, a targeting component may be coupled to a recombinant L chain or to a recombinant fusion LHN. The linkages between the botulinum components and the targeting moieties may include appropriate spacer components, which may also be DNA encoded.
In one embodiment, an LHN, which may be a hybrid of an L chain and an HN from different botulinum toxin types, is expressed recombinantly as a fusion protein. Such an LHN hybrid may also be coupled to a targeting component. There may be included one or more spacers between the L and HN and/or between the LHN and targeting component.
In another embodiment of the invention, the L chain of a botulinum neurotoxin, or a fragment of the L chain containing the endopeptidase activity, is expressed recombinantly to produce an agent for use in accordance with the present invention.
In another embodiment of the invention, the L chain of a botulinum neurotoxin, or a fragment of the L chain containing the endopeptidase activity, is expressed recombinantly as a fusion protein, with the HN of the H chain and the targeting component. The expressed fusion protein may also include one or more spacer regions. For example, the L chain may be fused to HN which is in turn fused to the targeting component. In another example, the HN may be fused to the L chain which is in turn fused to the targeting component. Spacer components may be expressed recombinantly between some or all of the components of an agent of the invention.
In one example of producing a hybrid of LHN, the L chain is obtained from botulinum toxin type B and the amine end segment of the HN chain fragment is obtained from botulinum toxin type A. The HN fragment of the botulinum toxin type A is produced according to the method described by Shone C. C., Hambleton, P., and Melling, J. (1987, Eur. J. Biochem. 167, 175-180) and the L chain of botulinum toxin type B according to the method of Sathyamoorthy, V. and DasGupta, B. R. (1985, J. Biol. Chem. 260, 10461-10466). The free cysteine on the amine end segment of the H chain fragment of botulinum toxin type A is then derivatized by the addition of a ten-fold molar excess of dipyridyl disulphide followed by incubation at 4° C. overnight. The excess dipvridyl disulphide and the thiopyridone by product are then removed by desalting the protein over a PD10 column (Pharmacia) into PBS.
The derivatized HN is then concentrated to a protein concentration in excess of 1 mg/ml before being mixed with an equimolar portion of L chain from botulinum toxin type B (>1 mg/ml in PBS). After overnight incubation at room temperature the mixture is separated by size exclusion chromatography over Superose 6 (Pharmacia), and the fractions analyzed by SDS-PAGE. The chimeric LHN is then available to produce a conjugated agent which includes a targeting component.
The example described above is purely illustrative of the invention. In synthesizing the agents, the coupling of the targeting moieties to the botulinum components, for example the modified botulinum neurotoxins or fragments thereof, may be achieved via chemical coupling using reagents and techniques known to those skilled in the art. Thus, any coupling chemistry capable of covalently attaching the targeting moieties of the agents to botulinum neurotoxin components and known to those skilled in the art is covered by the scope of this application.
Modified botulinum toxins which have an altered biological persistence and/or biological activity are contemplated for use in the present invention. U.S. patent application Ser. Nos. 09/620,840 and 09/910,346 include examples of compositions and methods for altering the biological persistence of botulinum toxins. These two patent applications are incorporated in their entirety herein by reference.
A biological persistence enhancing component and/or a biological activity enhancing component, for example, a leucine based motif, may be added to a botulinum neurotoxin thereby increasing the biological persistence and/or biological activity of the botulinum neurotoxin. Similarly, a biological persistence enhancing component can be removed from a botulinum neurotoxin thereby decreasing the biological persistence and/or biological activity of the neurotoxin.
The botulinum neurotoxin can be a hybrid neurotoxin. For example, the neurotoxin's targeting, translocation and therapeutic components may be derived from different botulinum toxin serotypes. For example, the polypeptide may comprise a first amino acid sequence region derived from the HC of a botulinum toxin type A, a second amino acid sequence region derived from the HN Of botulinum type B, and a third amino acid sequence region derived from the light chain of botulinum serotype E. This is merely an example and all other possible combinations are included within the scope of the present invention.
The neurotoxin's targeting, translocation and therapeutic components can be modified from the naturally occurring sequence from which they are derived. For example, the amino acid sequence region can have at least one or more amino acids added, deleted or substituted as compared to the naturally occurring sequence.
Amino acids that can be substituted for amino acids contained in a biological persistence enhancing component include alanine, aspargine, cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, proline, glutamine, arginine, serine, threonine, valine, tryptophan, tyrosine and other naturally occurring amino acids as well as non-standard amino acids.
Methods of the present invention provide for reduction in restenosis ranging from about 1% to about 100% in effectiveness. For example, the reduction may be between about 1% and about 10% or between about 10% and about 20% or between about 10% and about 30% or between about 10% and about 40% or between about 10% and about 50% or between about 10% and about 60% or between about 10% and about 70% or between about 10% and about 80% or between about 10% and about 90% or between about 10% and about 100%.
Generally, the dose of neurotoxin to be administered may vary with the age, presenting condition and weight of the patient to be treated. The potency of the neurotoxin will also be considered. Toxin potency is expressed as a multiple of the LD50 value for a mouse. One “unit” of toxin can be defined as the amount of toxin that kills 50% of a group of mice that were disease-free prior to inoculation with the toxin. For example, commercially available Botulinum toxin A typically has a potency such that one nanogram contains about 40 mouse units. The potency, or LD50 in humans of the Botulinum toxin A product supplied by Allergan, Inc. under the registered trademark “BOTOX” is believed to be about 2,730 mouse units.
The neurotoxin can be administered in a dose of about 0.001 units up to about 100 units. In one embodiment, individual dosages of about 0.01 units to about 5 units are used. In another embodiment, individual dosages of about 0.01 units to about 3 units are used. In still another embodiment, individual dosages of about 0.01 units to about 1 unit are used. In still another embodiment, individual dosages of about 0.05 units to about 1 unit are used. Those of ordinary skill in the art will know, or can readily ascertain, how to adjust dosages for neurotoxin of greater or lesser potency in a certain circumstance.
For modified or variant botulinum toxins potency may be expressed as a multiple of the LD50 value of an agent of the invention for a mouse. A “U” or “unit” of an agent can be defined as the amount of toxin that kills 50% of a group of mice that were disease-free prior to inoculation with the agent. Alternatively, potency may be expressed as the LD50 value of an agent that would be produced by an equal molar amount of a native, non-variant botulinum toxin.
Preferably, the lowest therapeutically effective dosage will be administered to the patient. The lowest therapeutic dosage is that dosage which results in the desired effect on a blood vessel of the patient to which the toxin is administered. Methods for assessing or quantifying the effect of a toxin on a blood vessel can be determined by those skilled in the art. For example, use of optical coherence topography can provide a measure of the dilating effect of the toxin.
In an initial treatment, a low dosage may be administered to determine the patient's sensitivity to, and tolerance of, the neurotoxin. Additional administrations of the same or different dosages may be administered to the blood vessel as necessary. For example, a toxin may be administered to a blood vessel before a procedure, for example, a coronary angioplasty procedure, is performed, and/or during the procedure and/or after the procedure. The number of administrations and timing of the administrations may be determined by the treating physician.
The neurotoxins may be administered by, for example, injection into a blood vessel using a needle or by needleless injection. The toxin may also be administered by application of the toxin to the wall of the blood vessel in a salve, lotion, ointment, cream, emulsion or the like delivery substrates.
In one embodiment, an agent of the invention is administered to the patient by injection. For example, an agent may be injected into the cardiovascular system of a patient. In particular, the injecting may be into an artery, for example, a coronary artery. Typically, the injecting is into the region of the blood vessel where the vessel is to undergo a procedure, for example, a procedure to mechanically expand the internal diameter of an occluded blood vessel.
An agent may be injected into the wall of a blood vessel from outside of the blood vessel or an agent may be injected into the wall of a blood vessel from inside the blood vessel. Methods for injecting into a wall of a blood vessel are well known to those of ordinary skill in the art. For example, an agent may be injected into the wall of a blood vessel from inside the blood vessel by the use of a catheter containing one or more needles for injecting.
In needleless injection delivery methods, microprojectile drug particles may be coated with a neurotoxin and then discharged into the blood vessel from an external delivery device. Depending on the discharge velocity and the distance from the injection site, the drug particles penetrate through the different layers of the blood vessels. As the microprojectiles penetrate through, or are deposited in, the blood vessel cells, the neurotoxin is released. Individual layers of blood vessels may be targeted for the microprojectiles.
The neurotoxins may also be administered using a stent or an angioplasty balloon that is coated or impregnated with the toxin, for example botulinum toxin. U.S. Pat. Nos. 6,306,423 and 6,312,708 disclose material that may be impregnated, attached or imbedded with the toxin and which may be used to coat stents and/or angioplasty balloons. In addition, the material may be used to form stents which include the toxin. The disclosure of each of these two patents is incorporated in its entirety herein by reference. In one embodiment, the blood vessel to be treated is first administered with botulinum toxin before inserting the toxin comprising stent and/or balloon. In another embodiment, the blood vessel to be treated is not administered with toxin before inserting the botulinum toxin comprising stent and/or balloon.
Administrations may be repeated if necessary. As a general guideline, botulinum toxin A administered into a blood vessel may produce a dilating and/or anti-inflammatory effect for, for example about 1 month to about 3 months, or for example, about 3 to about 6 months or for example from about 6 months to about 1 year.
The agent may be allowed to induce its effect on a blood vessel before a procedure, for example, a coronary angioplasty procedure, is performed. For example, the agent may be allowed to dilate the blood vessel and/or prevent inflammation of the blood vessel before a procedure is begun. A physician of ordinary skill can determine when the agent has exerted its effect(s) on a blood vessel.
The invention having been fully described, examples illustrating its practice are set forth below. These examples should not, however, be considered to limit the scope of the invention, which is defined by the appended claims.
EXAMPLES Example 1
Use of Botulinum Toxin in Balloon Angioplasty Where a Stent is Not Used
A 54 year old male patient complains of chest pains in an emergency room examination. The patient smokes 2 to 3 packs of cigarettes a day, is of average weight and has a family history of coronary arterial blockage. The patient is diagnosed as having an occlusion of the left coronary artery. A coronary angiogram is used to measure the narrowing of the arteries. It is estimated that the patient suffers from an 80% occlusion of the left coronary artery. He is scheduled for a balloon angioplasty procedure for the following week.
The physician begins the procedure by injecting between about 0.05 units and about 5 units of botulinum toxin type A into the wall of the left coronary artery of the patient. Following injection, the artery is allowed to dilate. A 3-millimeter noncompliant balloon catheter is then inserted into the femoral artery of the patient through the groin/upper thigh area. The catheter is then fed through the artery up into the heart using a video monitor to guide the process. A guide wire is advanced to the location of the blocked artery, and the balloon catheter is passed along the guide wire into the target area of coronary blockage. When the catheter reaches the target area, the balloon is inflated for a period of several seconds to several minutes. After deflation of the balloon, the same area may be treated with one or more additional inflations. Examination reveals little or no damage to the treated artery.
One year after the procedure there is no sign of restenosis and the patient appears in good health.
Example 2
Use of Botulinum Toxin in Balloon Angioplasty Where a Stent is Used
A 62 year old male patient who is approximately 30% overweight and has a serum cholesterol level of approximately 260 complains of chest pains. The patient is diagnosed as having coronary artery blockage and is scheduled for a percutaneous transluminal coronary angioplasty procedure.
Between about 0.01 units to about 1 unit of botulinum toxin is injected directly into the wall of the artery in the area of the blockage. Following injection, the artery is allowed to dilate. A 3-millimeter compliant balloon catheter and stent are then inserted into the interosseous artery of the patient through the wrist area. The catheter and stent are then fed through the interosseous artery to the area of blockage. A guide wire is advanced to the location of the blocked artery, and the catheter and stent are passed along the guide wire into the target area of coronary blockage. When the catheter reaches the target area, the balloon is inflated and the stent is correspondingly expanded bracing open the artery. The balloon is deflated and removed leaving in place the expanded stent. There is no sign of damage to the artery.
Six months after the procedure there is no sign of restenosis and the patient appears in good health.
Example 3
Use of Botulinum Toxin in Balloon Angioplasty With a Stent to Treat an Advanced Case of Restenosis
A 49 year old male patient is diagnosed with coronary arterial blockage as a result of restenosis. The patient has a history of coronary arterial blockage and has previously undergone a balloon angioplasty procedure. Six months after the procedure, the patient is diagnosed with an advanced case of restenosis.
The patient undergoes a percutaneous transluminal coronary angioplasty procedure in which botulinum toxin type A, B, C, D, D, F and/or G is used. Between about 0.1 units and about 4 units of botulinum toxin is injected into the wall of the blood vessel in the area of restenosis. Following injection, the artery is allowed to dilate. A 4-millimeter compliant balloon catheter and stent are then inserted into the femoral artery of the patient. The catheter and stent are then fed through the artery to the area of blockage using a video monitor to guide the process. A guide wire is advanced to the location of the blocked artery, and the catheter and stent are passed along the guide wire into the target area of coronary blockage. When the catheter and stent reach the target area, the balloon is inflated and the stent correspondingly expanded holding open the artery. The balloon is deflated and removed leaving in place the expanded stent. 3 to 6 months after the procedure, the blood vessel is re-injected with the botulinum toxin in the area of the stent.
Two years after the procedure there is no sign of restenosis and the patient appears in good health.
Example 4
Balloon Angioplasty Where a Stent Impregnated With Botulinum Toxin is Used
A 58 year old female patient is diagnosed with coronary arterial blockage. The patient is scheduled for a percutaneous transluminal coronary angioplasty procedure in which a stent impregnated with botulinum toxin type A, B, C, D, E, F and/or G is used.
Between about 0.1 units and about 2 units of a botulinum toxin is injected into the wall of the blood vessel in the area of blockage. Following injection, the artery is allowed to dilate. A 2-millimeter compliant balloon catheter and stent, which is coated or impregnated with a botulinum toxin, are then inserted into the femoral artery of the patient. The catheter and stent are passed through the femoral artery to the area of blockage using a video monitor to guide the process. A guide wire is advanced to the location of the blocked artery, and the catheter and stent are passed along the guide wire into the target area of coronary blockage. When the catheter and stent reach the target area, the balloon is inflated and the stent is correspondingly expanded bracing open the artery. The balloon is deflated and removed leaving in place the expanded stent.
One year after the procedure there is no sign of restenosis and the patient appears in good health.
Example 5
Balloon Angioplasty Where a Self Expanding Stent Impregnated With Botulinum Toxin is Used
A 50 year old male patient is diagnosed with coronary arterial blockage of the left coronary artery and is scheduled for a percutaneous transluminal coronary angioplasty procedure in which a self expanding stent impregnated with a botulinum toxin is used.
The physician begins the procedure by injecting between about 0.1 units and about 5 units of botulinum toxin type A into the wall of the left coronary artery of the patient. Following injection, the artery is allowed to dilate. A self expanding stent impregnated with the botulinum toxin is then inserted with a catheter into the common interosseous artery of the patient through the wrist area. The catheter and stent are passed through the interosseous artery to the area of blockage. A guide wire is advanced to the location of the blocked artery advancing the botulinum toxin impregnated, self expanding stent into the target area of coronary blockage. When the catheter reaches the target area, the stent is expanded bracing open the artery.
Two years after the procedure there is no sign of restenosis and the patient appears in good health.
Example 6
Injection of Botulinum Toxin by Use of a Catheter Injecting System
A 51 year old female patient complains of chest pains. The patient is overweight and has a serum cholesterol level of approximately 270. The patient is diagnosed as having coronary artery blockage. A coronary angiogram is used to measure the narrowing of the arteries. It is estimated that the patient suffers from a 70% to 90% occlusion of a coronary artery. She is scheduled for a percutaneous transluminal coronary angioplasty procedure.
Between bout 0.01 units and about 3 units of a botulinum toxin is injected directly into the wall of the artery in the area of the blockage. For injection, a catheter which includes one or more injection needles is inserted through the femoral artery of the patient through the groin/upper thigh area into the area of the coronary blockage. There the botulinum toxin is injected into the inner wall of the occluded blood vessel.
Following injection of the botulinum toxin, the artery is allowed to dilate. A 3-millimeter compliant balloon catheter and stent impregnated with botulinum toxin type A are then inserted into the femoral artery of the patient. The catheter and stent are fed through the femoral artery to the area of blockage using a video monitor to guide the process. A guide wire is advanced to the location of the blocked artery, and the catheter and stent is passed along the guide wire into the target area of coronary blockage. When the catheter reaches the target area, the balloon is inflated and the stent is correspondingly expanded bracing open the artery. The balloon is deflated and removed leaving in place the expanded stent. There is no sign of damage to the blood vessel.
One year after the procedure there is no sign of restenosis and the patient appears in good health.
My invention also includes within its scope the use of a neurotoxin, such as a botulinum toxin, in the preparation of a medicament for the treatment of a cardiovascular disease.
All references, articles, patents, applications and publications set forth above are incorporated herein by reference in their entireties.
Accordingly, the spirit and scope of the following claims should not be limited to the descriptions of the preferred embodiments set forth above.

Claims (14)

I claim:
1. A method for treating a cardiovascular disease the method comprising the steps of:
(a) administering an effective amount of a botulinum neurotoxin directly into a wall of a coronary artery of a patient with a coronary disease, and;
(b) carrying out upon the patient a coronary arterial cardiovascular procedure for reducing a coronary arterial blockage, thereby treating a cardiovascular disease.
2. The method of claim 1 wherein treating the cardiovascular disease prevents restenosis.
3. The method of claim 1 wherein the cardiovascular procedure includes angioplasty.
4. The method of claim 3 wherein the angioplasty does not include the step of inserting a stent into the blood vessel.
5. The method of claim 1 wherein the step of administering is accomplished using a stent coated or impregnated with the botulinum neurotoxin.
6. The method of claim 1 wherein the botulinum neurotoxin reduces or eliminates damage to a blood vessel.
7. The method of claim 6 wherein the botulinum neurotoxin reduces or eliminates damage to a blood vessel by dilating the blood vessel.
8. The method of claim 6 wherein the botulinum neurotoxin reduces or eliminates damage to a blood vessel by reducing or eliminating inflammation of the blood vessel.
9. The method of claim 1 wherein the botulinum neurotoxin is selected from the group consisting of botulinum neurotoxin types A, B, C, D, E, F, G mixtures thereof and combinations thereof.
10. The method of claim 1 wherein the botulinum neurotoxin is botulinum neurotoxin type A.
11. A method of preventing restenosis of a coronary artery, the method comprising the steps of:
(a) administering an effective amount of a botulinum neurotoxin into a wall of a coronary artery of a patient with a coronary disease, and;
(b) carrying out upon the patient a coronary arterial cardiovascular procedure for reducing a coronary arterial blockage, thereby preventing restenosis of the coronary artery.
12. The method of claim 11, wherein the botulinum neurotoxin is botulinum neurotoxin type A.
13. A composition for use in a coronary arterial cardiovascular procedure comprising a stent for reducing a coronary arterial blockage with a botulinum neurotoxin attached to or imbedded in the stent.
14. The composition of claim 13 wherein the botulinum neurotoxin is botulinum neurotoxin type A.
US10/114,740 1999-08-10 2002-04-01 Methods for treating cardiovascular diseases with botulinum toxin Expired - Fee Related US6767544B2 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
US10/114,740 US6767544B2 (en) 2002-04-01 2002-04-01 Methods for treating cardiovascular diseases with botulinum toxin
PCT/US2003/009157 WO2003084567A1 (en) 2002-04-01 2003-03-24 Use of botulinum toxin for treating cardiovasular diseases
BR0308928-2A BR0308928A (en) 2002-04-01 2003-03-24 Use of botulinum toxin for the treatment of cardiovascular disease, as well as composition for use in a cardiovascular procedure.
AT03716821T ATE355852T1 (en) 2002-04-01 2003-03-24 USE OF BOTULINUM TOXIN FOR THE TREATMENT OF HEART AND Circulatory Diseases
JP2003581806A JP2005521735A (en) 2002-04-01 2003-03-24 Use of botulinum toxin to treat cardiovascular disease
AU2003220511A AU2003220511C1 (en) 2002-04-01 2003-03-24 Use of botulinum toxin for treating cardiovascular diseases
EP03716821A EP1490097B2 (en) 2002-04-01 2003-03-24 Use of botulinum toxin for treating cardiovascular diseases
KR10-2004-7015481A KR20040105818A (en) 2002-04-01 2003-03-24 Methods for treating cardiovascular diseases with botulinum toxin
MXPA04009410A MXPA04009410A (en) 2002-04-01 2003-03-24 Use of botulinum toxin for treating cardiovasular diseases.
ES03716821T ES2281636T5 (en) 2002-04-01 2003-03-24 USE OF BOTULINIC TOXIN TO TREAT CARDIOVASCULAR DISEASES.
CA002480505A CA2480505A1 (en) 2002-04-01 2003-03-24 Use of botulinum toxin for treating cardiovasular diseases
DE60312309T DE60312309T3 (en) 2002-04-01 2003-03-24 USE OF BOTULINUM TOXIN TO TREAT HEART AND CIRCULAR DISEASES
US10/628,905 US7048927B2 (en) 1999-08-10 2003-07-28 Botulinum neurotoxin eluting stent
US10/870,603 US7494654B2 (en) 1999-08-10 2004-06-16 Compositions and methods for treating cardiovascular conditions with botulinum toxin
HK05103242A HK1070575A1 (en) 2002-04-01 2005-04-15 Use of botulinum toxin for treating cardiovasculardiseases
US11/345,736 US7223399B2 (en) 1999-08-10 2006-02-01 Methods for treating restenosis with a botulinum neurotoxin
US11/446,403 US20060216313A1 (en) 1999-08-10 2006-06-02 Methods for treating a stricture with a botulinum toxin
JP2009032291A JP2009102440A (en) 2002-04-01 2009-02-16 Use of botulinum toxin for treating cardiovascular diseases
US12/436,730 US20090220568A1 (en) 1999-08-10 2009-05-06 Methods for treating a stricture with a botulinum toxin
JP2012277214A JP2013053167A (en) 2002-04-01 2012-12-19 Use of botulinum toxin for treating cardiovascular disease

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/114,740 US6767544B2 (en) 2002-04-01 2002-04-01 Methods for treating cardiovascular diseases with botulinum toxin

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US09/371,354 Continuation-In-Part US6977080B1 (en) 1999-08-10 1999-08-10 Intrapericardial botulinum toxin treatment for bradycardia
US09/371,354 Continuation US6977080B1 (en) 1999-08-10 1999-08-10 Intrapericardial botulinum toxin treatment for bradycardia
US09/371,352 Continuation-In-Part US6263040B1 (en) 1999-08-10 1999-08-10 Methods and apparatus for cone-tilted parallel sampling and reconstruction

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/628,905 Continuation US7048927B2 (en) 1999-08-10 2003-07-28 Botulinum neurotoxin eluting stent

Publications (2)

Publication Number Publication Date
US20030185860A1 US20030185860A1 (en) 2003-10-02
US6767544B2 true US6767544B2 (en) 2004-07-27

Family

ID=28453838

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/114,740 Expired - Fee Related US6767544B2 (en) 1999-08-10 2002-04-01 Methods for treating cardiovascular diseases with botulinum toxin
US10/628,905 Expired - Fee Related US7048927B2 (en) 1999-08-10 2003-07-28 Botulinum neurotoxin eluting stent
US10/870,603 Expired - Fee Related US7494654B2 (en) 1999-08-10 2004-06-16 Compositions and methods for treating cardiovascular conditions with botulinum toxin
US11/345,736 Expired - Fee Related US7223399B2 (en) 1999-08-10 2006-02-01 Methods for treating restenosis with a botulinum neurotoxin

Family Applications After (3)

Application Number Title Priority Date Filing Date
US10/628,905 Expired - Fee Related US7048927B2 (en) 1999-08-10 2003-07-28 Botulinum neurotoxin eluting stent
US10/870,603 Expired - Fee Related US7494654B2 (en) 1999-08-10 2004-06-16 Compositions and methods for treating cardiovascular conditions with botulinum toxin
US11/345,736 Expired - Fee Related US7223399B2 (en) 1999-08-10 2006-02-01 Methods for treating restenosis with a botulinum neurotoxin

Country Status (13)

Country Link
US (4) US6767544B2 (en)
EP (1) EP1490097B2 (en)
JP (3) JP2005521735A (en)
KR (1) KR20040105818A (en)
AT (1) ATE355852T1 (en)
AU (1) AU2003220511C1 (en)
BR (1) BR0308928A (en)
CA (1) CA2480505A1 (en)
DE (1) DE60312309T3 (en)
ES (1) ES2281636T5 (en)
HK (1) HK1070575A1 (en)
MX (1) MXPA04009410A (en)
WO (1) WO2003084567A1 (en)

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030130722A1 (en) * 2001-12-21 2003-07-10 Marx Steven O. C3 exoenzyme-coated stents and uses thereof for treating and preventing restenosis
US20030211975A1 (en) * 2000-05-01 2003-11-13 Imarx Therapeutics Inc Method and apparatus for vascular neuromuscular blockade
US20040067235A1 (en) * 2002-07-29 2004-04-08 Rajiv Doshi Methods for the use of neurotoxin in the treatment of urologic disorders
US20040142005A1 (en) * 1999-08-10 2004-07-22 Allergan, Inc. Botulinum toxin eluting stent
US20050152923A1 (en) * 2004-01-08 2005-07-14 Brin Mitchell F. Methods for treating vascular disorders
US6977080B1 (en) * 1999-08-10 2005-12-20 Allergan, Inc. Intrapericardial botulinum toxin treatment for bradycardia
US20060024331A1 (en) * 2004-08-02 2006-02-02 Ester Fernandez-Salas Toxin compounds with enhanced membrane translocation characteristics
US20060062808A1 (en) * 2004-09-18 2006-03-23 Asthmatx, Inc. Inactivation of smooth muscle tissue
US20060211619A1 (en) * 2005-03-15 2006-09-21 Steward Lance E Multivalent clostridial toxin derivatives and methods of their use
US20060216313A1 (en) * 1999-08-10 2006-09-28 Allergan, Inc. Methods for treating a stricture with a botulinum toxin
US20060225742A1 (en) * 2003-05-13 2006-10-12 The Foundry, Inc. Apparatus for treating asthma using neurotoxin
US20070025919A1 (en) * 2005-07-22 2007-02-01 The Foundry Inc. Systems and methods for delivery of a therapeutic agent
US20070259002A1 (en) * 2004-10-29 2007-11-08 John Batchelor Botulinum Toxin Therapy of Heart Rhythm Disorders
US20070267011A1 (en) * 2006-05-19 2007-11-22 The Foundry Inc. Apparatus for toxin delivery to the nasal cavity
US20080004662A1 (en) * 2003-11-13 2008-01-03 Peters Nicholas S Post-operative control of cardiac arrhythmia by modification of neuronal signaling through fat pads of the heart
US20080092910A1 (en) * 2006-10-18 2008-04-24 Allergan, Inc. Apparatus and method for treating obesity using neurotoxins in conjunction with bariatric procedures
EP1982997A1 (en) 2004-09-01 2008-10-22 Allergan, Inc. Degradable clostridial toxins
US20090028908A1 (en) * 2002-02-25 2009-01-29 Allergan, Inc. Methods for treating neurogenic inflammation
US20090048431A1 (en) * 2004-06-30 2009-02-19 Allergan, Inc. Multivalent clostridial toxins
US20090232850A1 (en) * 2008-03-13 2009-09-17 Manack Aubrey N Therapeutic treatments using botulinum neurotoxin
US7794386B2 (en) 2006-03-15 2010-09-14 Allergan, Inc. Methods for facilitating weight loss
EP2316847A2 (en) 2005-03-15 2011-05-04 Allergan, Inc. Modified clostridial toxins with enhanced targeting capabilities for endogenous clostridial toxin receptor systems
US8088127B2 (en) 2008-05-09 2012-01-03 Innovative Pulmonary Solutions, Inc. Systems, assemblies, and methods for treating a bronchial tree
US20120172837A1 (en) * 2002-04-08 2012-07-05 Ardian, Inc. Methods for inhibiting renal nerve activity via an intra-to-extravascular approach
US8483831B1 (en) 2008-02-15 2013-07-09 Holaira, Inc. System and method for bronchial dilation
WO2014053651A1 (en) 2012-10-04 2014-04-10 Dublin City University Biotherapy for pain
US8740895B2 (en) 2009-10-27 2014-06-03 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US8768451B2 (en) 2010-04-30 2014-07-01 Boston Scientific Scimed, Inc. Therapeutic agent delivery device for delivery of a neurotoxin
US8771252B2 (en) 2002-04-08 2014-07-08 Medtronic Ardian Luxembourg S.A.R.L. Methods and devices for renal nerve blocking
US8852163B2 (en) 2002-04-08 2014-10-07 Medtronic Ardian Luxembourg S.A.R.L. Renal neuromodulation via drugs and neuromodulatory agents and associated systems and methods
US8911439B2 (en) 2009-11-11 2014-12-16 Holaira, Inc. Non-invasive and minimally invasive denervation methods and systems for performing the same
US9149328B2 (en) 2009-11-11 2015-10-06 Holaira, Inc. Systems, apparatuses, and methods for treating tissue and controlling stenosis
US20150282877A1 (en) * 2002-04-08 2015-10-08 Medtronic Ardian Luxembourg S.A.R.L. Methods and apparatus for thermally-induced renal neuromodulation
US9192715B2 (en) 2002-04-08 2015-11-24 Medtronic Ardian Luxembourg S.A.R.L. Methods for renal nerve blocking
US9265558B2 (en) 2002-04-08 2016-02-23 Medtronic Ardian Luxembourg S.A.R.L. Methods for bilateral renal neuromodulation
US9398933B2 (en) 2012-12-27 2016-07-26 Holaira, Inc. Methods for improving drug efficacy including a combination of drug administration and nerve modulation
US9636174B2 (en) 2002-04-08 2017-05-02 Medtronic Ardian Luxembourg S.A.R.L. Methods for therapeutic renal neuromodulation
US9713483B2 (en) 1995-10-13 2017-07-25 Medtronic Vascular, Inc. Catheters and related devices for forming passageways between blood vessels or other anatomical structures
US20170246268A1 (en) * 2014-09-12 2017-08-31 Gary E.l BORODIC Longevity Enhancement Induced by Repetitive Injections of Botulinum Toxin (Centurion Effect)
WO2018002348A1 (en) 2016-07-01 2018-01-04 Ipsen Biopharm Limited Production of activated clostridial neurotoxins
US9920310B2 (en) 2013-07-09 2018-03-20 Ipsen Bioinnovation, Limited Cationic neurotoxins
US9919144B2 (en) 2011-04-08 2018-03-20 Medtronic Adrian Luxembourg S.a.r.l. Iontophoresis drug delivery system and method for denervation of the renal sympathetic nerve and iontophoretic drug delivery
WO2018060351A1 (en) 2016-09-29 2018-04-05 Ipsen Biopharm Limited Hybrid neurotoxins
US10052465B2 (en) 2005-07-22 2018-08-21 The Foundry, Llc Methods and systems for toxin delivery to the nasal cavity
US10130792B2 (en) 2002-04-08 2018-11-20 Medtronic Ardian Luxembourg S.A.R.L. Methods for therapeutic renal neuromodulation using neuromodulatory agents or drugs
US10149893B2 (en) 2013-09-24 2018-12-11 Allergan, Inc. Methods for modifying progression of osteoarthritis
US10350004B2 (en) 2004-12-09 2019-07-16 Twelve, Inc. Intravascular treatment catheters
WO2019162696A1 (en) 2018-02-26 2019-08-29 Ipsen Biopharm Limited Use of ultrasound to guide injection of non-cytotoxic protease
US10457927B2 (en) 2013-12-23 2019-10-29 Dublin City University Multiprotease therapeutics for chronic pain
EP3650462A1 (en) 2015-10-02 2020-05-13 Ipsen Biopharm Limited Method for purifying clostridial neurotoxin
EP3822286A1 (en) 2015-01-09 2021-05-19 Ipsen Bioinnovation Limited Cationic neurotoxins
US11116561B2 (en) 2018-01-24 2021-09-14 Medtronic Ardian Luxembourg S.A.R.L. Devices, agents, and associated methods for selective modulation of renal nerves
EP3889168A1 (en) 2016-05-05 2021-10-06 Ipsen Biopharm Limited Chimeric neurotoxins
WO2022189807A2 (en) 2021-03-11 2022-09-15 Ipsen Biopharm Limited Modified clostridial neurotoxins

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9833618B2 (en) * 2005-02-04 2017-12-05 Palo Alto Investors Methods and compositions for treating a disease condition in a subject
EP2264458A1 (en) 2005-04-05 2010-12-22 Allergan, Inc. Clostridial toxin activity assays
US20070237802A1 (en) * 2006-04-11 2007-10-11 Medtronic Vascular, Inc. Inhibition of Calcification on an Endovascular Device
WO2010101968A1 (en) 2009-03-06 2010-09-10 Allergan, Inc. Clostridial toxin to improve ejaculate
US9125907B2 (en) * 2009-09-30 2015-09-08 Christopher Shaari Use of botulinum neurotoxin to treat substance addictions
US20120207743A1 (en) * 2011-02-14 2012-08-16 Allergan, Inc. Inhibiting Aberrant Blood Vessel Formation Using Retargeted Endopeptidases

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5092841A (en) 1990-05-17 1992-03-03 Wayne State University Method for treating an arterial wall injured during angioplasty
US5385935A (en) 1992-09-14 1995-01-31 Kissei Pharmaceutical Co., Ltd. Method for the inhibition of restenosis associated with coronary intervention
US5437291A (en) * 1993-08-26 1995-08-01 Univ Johns Hopkins Method for treating gastrointestinal muscle disorders and other smooth muscle dysfunction
US5562907A (en) * 1993-05-14 1996-10-08 Arnon; Stephen S. Method to prevent side-effects and insensitivity to the therapeutic uses of toxins
US5766605A (en) 1994-04-15 1998-06-16 Mount Sinai School Of Medicine Of The City University Of New York Treatment of autonomic nerve dysfunction with botulinum toxin
US5851786A (en) * 1995-09-27 1998-12-22 National Jewish Center For Immunology And Respiratory Medicine Product and process to regulate actin polymerization
US5939070A (en) * 1996-10-28 1999-08-17 Wisconsin Alumni Research Foundation Hybrid botulinal neurotoxins
US5989280A (en) 1993-10-22 1999-11-23 Scimed Lifesystems, Inc Stent delivery apparatus and method
US6057367A (en) * 1996-08-30 2000-05-02 Duke University Manipulating nitrosative stress to kill pathologic microbes, pathologic helminths and pathologically proliferating cells or to upregulate nitrosative stress defenses
US6063768A (en) * 1997-09-04 2000-05-16 First; Eric R. Application of botulinum toxin to the management of neurogenic inflammatory disorders
US6102904A (en) * 1995-07-10 2000-08-15 Interventional Technologies, Inc. Device for injecting fluid into a wall of a blood vessel
US6183506B1 (en) 1996-03-05 2001-02-06 Divysio Solutions Ltd. Expandable stent and method for delivery of same
WO2001010458A1 (en) * 1999-08-10 2001-02-15 Allergan Sales, Inc. Use of neurotoxin for treating cardiac muscle disorders
US6190404B1 (en) 1997-11-07 2001-02-20 Advanced Bio Prosthetic Surfaces, Ltd. Intravascular stent and method for manufacturing an intravascular stent
US6196230B1 (en) 1998-09-10 2001-03-06 Percardia, Inc. Stent delivery system and method of use
US6239118B1 (en) 1999-10-05 2001-05-29 Richard A. Schatz Method for preventing restenosis using a substituted adenine derivative
US6238872B1 (en) * 1997-04-01 2001-05-29 S.E.T.-Smart Endolumenal Technologies Ltd. Targeted therapy to a biomedical device
US6241758B1 (en) 1999-05-28 2001-06-05 Advanced Cardiovascular Systems, Inc. Self-expanding stent delivery system and method of use
US6261318B1 (en) 1995-07-25 2001-07-17 Medstent Inc. Expandable stent
US6264671B1 (en) 1999-11-15 2001-07-24 Advanced Cardiovascular Systems, Inc. Stent delivery catheter and method of use
US6270521B1 (en) 1999-05-21 2001-08-07 Cordis Corporation Stent delivery catheter system for primary stenting
US6290961B1 (en) * 1993-12-28 2001-09-18 Allergan, Inc. Method for treating dystonia with botulinum toxin type B
US6293959B1 (en) 1998-11-16 2001-09-25 Cordis Corporation Balloon catheter and stent delivery system having enhanced stent retention and method
US6299893B1 (en) * 2000-04-17 2001-10-09 Marvin Schwartz Method to reduce hair loss and stimulate hair regrowth
US6306162B1 (en) 1999-12-15 2001-10-23 Advanced Cardiovascular Systems, Inc. Stent delivery system utilizing novel balloon for obtaining variable post-deployment stent characteristics
US6306423B1 (en) 2000-06-02 2001-10-23 Allergan Sales, Inc. Neurotoxin implant
WO2001082947A1 (en) * 2000-05-01 2001-11-08 Unger Evan C Method and apparatus for vascular neuromuscular blockade
US6344055B1 (en) 1997-05-14 2002-02-05 Novo Rps Ulc Method for production of an expandable stent
US6429189B1 (en) * 1999-12-10 2002-08-06 Botulinum Toxin Research Associates, Inc. Cytotoxin (non-neurotoxin) for the treatment of human headache disorders and inflammatory diseases
US6464986B1 (en) 2000-04-14 2002-10-15 Allegan Sales, Inc. Method for treating pain by peripheral administration of a neurotoxin

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993020784A1 (en) 1992-04-10 1993-10-28 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education On Behalf Of The Oregon Health Sciences University A microneedle for injection of ocular blood vessels
US6121296A (en) * 1992-11-04 2000-09-19 Albert Einstein College Of Medicine Of Yeshiva University Transition-state inhibitors for nucleoside hydrolase and transferase reactions
EP1602379A1 (en) * 1993-12-28 2005-12-07 Allergan, Inc. Botulinum toxin B for treating spastic muscle
CA2190011C (en) * 1994-05-09 2000-09-26 William J. Binder Method for reduction of headache pain
US5626562A (en) 1994-11-28 1997-05-06 Devices For Vascular Intervention Drug delivery catheter
CA2338093C (en) 1998-07-20 2010-11-30 Shousun C. Szu Vaccines against escherichia coli o157 infection
US6767544B2 (en) * 2002-04-01 2004-07-27 Allergan, Inc. Methods for treating cardiovascular diseases with botulinum toxin
GB9922554D0 (en) 1999-09-23 1999-11-24 Microbiological Res Authority Inhibition of secretion from non-neuronal cells
JP2004516333A (en) 2001-01-04 2004-06-03 ユニバーシティー オブ サスカチェワン Enterohemorrhagic Escherichia coli vaccine
US20020099356A1 (en) * 2001-01-19 2002-07-25 Unger Evan C. Transmembrane transport apparatus and method

Patent Citations (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5092841A (en) 1990-05-17 1992-03-03 Wayne State University Method for treating an arterial wall injured during angioplasty
US5385935A (en) 1992-09-14 1995-01-31 Kissei Pharmaceutical Co., Ltd. Method for the inhibition of restenosis associated with coronary intervention
US5562907A (en) * 1993-05-14 1996-10-08 Arnon; Stephen S. Method to prevent side-effects and insensitivity to the therapeutic uses of toxins
US5437291A (en) * 1993-08-26 1995-08-01 Univ Johns Hopkins Method for treating gastrointestinal muscle disorders and other smooth muscle dysfunction
US5989280A (en) 1993-10-22 1999-11-23 Scimed Lifesystems, Inc Stent delivery apparatus and method
US6290961B1 (en) * 1993-12-28 2001-09-18 Allergan, Inc. Method for treating dystonia with botulinum toxin type B
US6319505B1 (en) * 1993-12-28 2001-11-20 Allergan Sales, Inc. Method for treating dystonia with botulinum toxin types C to G
US5766605A (en) 1994-04-15 1998-06-16 Mount Sinai School Of Medicine Of The City University Of New York Treatment of autonomic nerve dysfunction with botulinum toxin
US6102904A (en) * 1995-07-10 2000-08-15 Interventional Technologies, Inc. Device for injecting fluid into a wall of a blood vessel
US6261318B1 (en) 1995-07-25 2001-07-17 Medstent Inc. Expandable stent
US5851786A (en) * 1995-09-27 1998-12-22 National Jewish Center For Immunology And Respiratory Medicine Product and process to regulate actin polymerization
US6217608B1 (en) 1996-03-05 2001-04-17 Divysio Solutions Ulc Expandable stent and method for delivery of same
US6183506B1 (en) 1996-03-05 2001-02-06 Divysio Solutions Ltd. Expandable stent and method for delivery of same
US6057367A (en) * 1996-08-30 2000-05-02 Duke University Manipulating nitrosative stress to kill pathologic microbes, pathologic helminths and pathologically proliferating cells or to upregulate nitrosative stress defenses
US5939070A (en) * 1996-10-28 1999-08-17 Wisconsin Alumni Research Foundation Hybrid botulinal neurotoxins
US6238872B1 (en) * 1997-04-01 2001-05-29 S.E.T.-Smart Endolumenal Technologies Ltd. Targeted therapy to a biomedical device
US6344055B1 (en) 1997-05-14 2002-02-05 Novo Rps Ulc Method for production of an expandable stent
US6063768A (en) * 1997-09-04 2000-05-16 First; Eric R. Application of botulinum toxin to the management of neurogenic inflammatory disorders
US6190404B1 (en) 1997-11-07 2001-02-20 Advanced Bio Prosthetic Surfaces, Ltd. Intravascular stent and method for manufacturing an intravascular stent
US6196230B1 (en) 1998-09-10 2001-03-06 Percardia, Inc. Stent delivery system and method of use
US6293959B1 (en) 1998-11-16 2001-09-25 Cordis Corporation Balloon catheter and stent delivery system having enhanced stent retention and method
US6270521B1 (en) 1999-05-21 2001-08-07 Cordis Corporation Stent delivery catheter system for primary stenting
US6241758B1 (en) 1999-05-28 2001-06-05 Advanced Cardiovascular Systems, Inc. Self-expanding stent delivery system and method of use
WO2001010458A1 (en) * 1999-08-10 2001-02-15 Allergan Sales, Inc. Use of neurotoxin for treating cardiac muscle disorders
US6239118B1 (en) 1999-10-05 2001-05-29 Richard A. Schatz Method for preventing restenosis using a substituted adenine derivative
US6264671B1 (en) 1999-11-15 2001-07-24 Advanced Cardiovascular Systems, Inc. Stent delivery catheter and method of use
US6429189B1 (en) * 1999-12-10 2002-08-06 Botulinum Toxin Research Associates, Inc. Cytotoxin (non-neurotoxin) for the treatment of human headache disorders and inflammatory diseases
US6306162B1 (en) 1999-12-15 2001-10-23 Advanced Cardiovascular Systems, Inc. Stent delivery system utilizing novel balloon for obtaining variable post-deployment stent characteristics
US6464986B1 (en) 2000-04-14 2002-10-15 Allegan Sales, Inc. Method for treating pain by peripheral administration of a neurotoxin
US6299893B1 (en) * 2000-04-17 2001-10-09 Marvin Schwartz Method to reduce hair loss and stimulate hair regrowth
WO2001082947A1 (en) * 2000-05-01 2001-11-08 Unger Evan C Method and apparatus for vascular neuromuscular blockade
US6579847B1 (en) * 2000-05-01 2003-06-17 Imarx Therapeutics Inc. Method and apparatus for vascular neuromuscular blockade
US6306423B1 (en) 2000-06-02 2001-10-23 Allergan Sales, Inc. Neurotoxin implant
US6312708B1 (en) 2000-06-02 2001-11-06 Allergan Sales, Inc. Botulinum toxin implant
US6383509B1 (en) * 2000-06-02 2002-05-07 Allergan Sales, Inc. Biodegradable neurotoxin implant

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
Aktories, K., et al., Clostridium botulinum C3 ADP-Ribosyltransferase, Current Topics in Microbiology and Immunology, vol. 175, 1992, 115-131.
Author Unknown, Internet, Drug-coated stents help eliminate angioplasty problems, Mar. 18, 2002, www.cnn.com/2002/helath/03/18/heart.angioplasty.ap/index.html. 3 pgs.
Claus, D., et al., Botulinum Toxin: influence on respiratory heart rate variation, Mov Disord Sep. 1995; 10(5) 574-579.
DePalma, GD et al, Endoscopy, Dec. 2001, vol. 33(12), pp. 1027-1030.* *
Fauci, A.S., et al., 14<th >Edition Harrison's Principles of Internal Medicine, McGraw-Hill, pp. 904-907, 1998.
Fauci, A.S., et al., 14th Edition Harrison's Principles of Internal Medicine, McGraw-Hill, pp. 904-907, 1998.
Fiorentini, C. et al., Bacterial toxins and the Rho GTP-binding protein: what microbes teach us about cell regulation, Cell Death and Differentiation (1998) 5, pp. 720-728.
Girlanda, P., et al., Botulinum toxin therapy: distant effects on neuromuscular transmission and autonomic nervous system, J. Neurol Neurosurg Psychiatry 1992;55(9):844-5.
Lamanna, C., et al., Cardiac Effects of Botulinal Toxin, Arch Int Pharmacodyn Ther 1988; 293: 69-83.
Malnick, S., et al., Fatal heart block following treatment with botulinum toxin for achalasia, Am J Gastroenterol 2000;Nov; 95(11):3333-3334.
Maria, G et al, Diseases of the colon & Rectum, 2000, vol. 43(5), May, pp. 721-723.* *
Muniyappa, R et al, Am. J. Physiol. Heart Cir. Physiol., vol. 278, (6) pp. H1762-H1768, Jun. 2000.* *
Nebe, A., et al., No effects on heart-rate variability and cardiovascular reflex tests after boulinum toxin treatment of cervical dystonia, Mov Disord May 1996; 11(3):337-339.
Nishiki, T., et al., Morphological effects of clostridium botulinum C3 exoenzyme on cultured cells, Jpn. J. Med. Sci. Biol. 43, 1990, 261-262.
Sathyamoorthy, V., et al., Separation, purification, particial characterization and comparison of the heavy and light chains of botulinum neurotoxin types A, B and E, The J. of Biol. Chem, vol. 260, No. 19, Sep. 5, pp. 10461-10466, 1985.
Shone, C., et al., A 50-kDa fragment from the NH2-terminus of the heavy subunit of Clostridium botulinumtype A neurotoxin forms channels in lipid vesicles, Eur. J. Biochem 167, 175-180, 1987.
Tan, EK et al, Movement disorders, vol. 14(2), pp. 345-349, 1999.* *

Cited By (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9713483B2 (en) 1995-10-13 2017-07-25 Medtronic Vascular, Inc. Catheters and related devices for forming passageways between blood vessels or other anatomical structures
US7494654B2 (en) * 1999-08-10 2009-02-24 Allergan, Inc. Compositions and methods for treating cardiovascular conditions with botulinum toxin
US20060083757A1 (en) * 1999-08-10 2006-04-20 Allergan, Inc. Method for treating cardiac muscle disorders
US20040142005A1 (en) * 1999-08-10 2004-07-22 Allergan, Inc. Botulinum toxin eluting stent
US20040223975A1 (en) * 1999-08-10 2004-11-11 Allergan, Inc. Methods for treating cardiovascular diseases with botulinum toxin
US20060216313A1 (en) * 1999-08-10 2006-09-28 Allergan, Inc. Methods for treating a stricture with a botulinum toxin
US20090220568A1 (en) * 1999-08-10 2009-09-03 Allergan, Inc. Methods for treating a stricture with a botulinum toxin
US20060127417A1 (en) * 1999-08-10 2006-06-15 Allergan, Inc. Methods for treating restenosis with a botulinum neurotoxin
US7485624B2 (en) * 1999-08-10 2009-02-03 Allergan, Inc. Method for treating cardiac muscle disorders
US6977080B1 (en) * 1999-08-10 2005-12-20 Allergan, Inc. Intrapericardial botulinum toxin treatment for bradycardia
US7048927B2 (en) * 1999-08-10 2006-05-23 Allergan, Inc. Botulinum neurotoxin eluting stent
US7223399B2 (en) * 1999-08-10 2007-05-29 Allergan, Inc. Methods for treating restenosis with a botulinum neurotoxin
US20030211975A1 (en) * 2000-05-01 2003-11-13 Imarx Therapeutics Inc Method and apparatus for vascular neuromuscular blockade
US7364586B2 (en) * 2001-12-21 2008-04-29 The Trustees Of Columbia University In The City Of New York C3 exoenzyme-coated stents and uses thereof for treating and preventing restenosis
US20030130722A1 (en) * 2001-12-21 2003-07-10 Marx Steven O. C3 exoenzyme-coated stents and uses thereof for treating and preventing restenosis
US7662178B2 (en) 2001-12-21 2010-02-16 The Trustees Of Columbia University In The City Of New York C3 exoenzyme-coated stents and uses thereof for treating and preventing restenosis
US20090028908A1 (en) * 2002-02-25 2009-01-29 Allergan, Inc. Methods for treating neurogenic inflammation
US20150282877A1 (en) * 2002-04-08 2015-10-08 Medtronic Ardian Luxembourg S.A.R.L. Methods and apparatus for thermally-induced renal neuromodulation
US8852163B2 (en) 2002-04-08 2014-10-07 Medtronic Ardian Luxembourg S.A.R.L. Renal neuromodulation via drugs and neuromodulatory agents and associated systems and methods
US10124195B2 (en) 2002-04-08 2018-11-13 Medtronic Ardian Luxembourg S.A.R.L. Methods for thermally-induced renal neuromodulation
US10130792B2 (en) 2002-04-08 2018-11-20 Medtronic Ardian Luxembourg S.A.R.L. Methods for therapeutic renal neuromodulation using neuromodulatory agents or drugs
US8728138B2 (en) 2002-04-08 2014-05-20 Medtronic Ardian Luxembourg S.A.R.L. Methods for thermally-induced renal neuromodulation
US9636174B2 (en) 2002-04-08 2017-05-02 Medtronic Ardian Luxembourg S.A.R.L. Methods for therapeutic renal neuromodulation
US9186198B2 (en) 2002-04-08 2015-11-17 Medtronic Ardian Luxembourg S.A.R.L. Ultrasound apparatuses for thermally-induced renal neuromodulation and associated systems and methods
US9968611B2 (en) 2002-04-08 2018-05-15 Medtronic Ardian Luxembourg S.A.R.L. Methods and devices for renal nerve blocking
US10179027B2 (en) 2002-04-08 2019-01-15 Medtronic Ardian Luxembourg S.A.R.L. Catheter apparatuses having expandable baskets for renal neuromodulation and associated systems and methods
US8728137B2 (en) 2002-04-08 2014-05-20 Medtronic Ardian Luxembourg S.A.R.L. Methods for thermally-induced renal neuromodulation
US8684998B2 (en) * 2002-04-08 2014-04-01 Medtronic Ardian Luxembourg S.A.R.L. Methods for inhibiting renal nerve activity
US8771252B2 (en) 2002-04-08 2014-07-08 Medtronic Ardian Luxembourg S.A.R.L. Methods and devices for renal nerve blocking
US8784463B2 (en) 2002-04-08 2014-07-22 Medtronic Ardian Luxembourg S.A.R.L. Methods for thermally-induced renal neuromodulation
US10850091B2 (en) 2002-04-08 2020-12-01 Medtronic Ardian Luxembourg S.A.R.L. Methods and apparatus for bilateral renal neuromodulation
US20120172837A1 (en) * 2002-04-08 2012-07-05 Ardian, Inc. Methods for inhibiting renal nerve activity via an intra-to-extravascular approach
US8845629B2 (en) 2002-04-08 2014-09-30 Medtronic Ardian Luxembourg S.A.R.L. Ultrasound apparatuses for thermally-induced renal neuromodulation
US9265558B2 (en) 2002-04-08 2016-02-23 Medtronic Ardian Luxembourg S.A.R.L. Methods for bilateral renal neuromodulation
US10034708B2 (en) * 2002-04-08 2018-07-31 Medtronic Ardian Luxembourg S.A.R.L. Methods and apparatus for thermally-induced renal neuromodulation
US9814873B2 (en) 2002-04-08 2017-11-14 Medtronic Ardian Luxembourg S.A.R.L. Methods and apparatus for bilateral renal neuromodulation
US10441356B2 (en) 2002-04-08 2019-10-15 Medtronic Ardian Luxembourg S.A.R.L. Methods for renal neuromodulation via neuromodulatory agents
US9192715B2 (en) 2002-04-08 2015-11-24 Medtronic Ardian Luxembourg S.A.R.L. Methods for renal nerve blocking
US10179235B2 (en) 2002-04-08 2019-01-15 Medtronic Ardian Luxembourg S.A.R.L. Methods and apparatus for bilateral renal neuromodulation
US10376516B2 (en) 2002-04-08 2019-08-13 Medtronic Ardian Luxembourg S.A.R.L. Methods and devices for renal nerve blocking
US10293190B2 (en) 2002-04-08 2019-05-21 Medtronic Ardian Luxembourg S.A.R.L. Thermally-induced renal neuromodulation and associated systems and methods
US8986294B2 (en) 2002-04-08 2015-03-24 Medtronic Ardian Luxembourg S.a.rl. Apparatuses for thermally-induced renal neuromodulation
US20040067235A1 (en) * 2002-07-29 2004-04-08 Rajiv Doshi Methods for the use of neurotoxin in the treatment of urologic disorders
US10953170B2 (en) 2003-05-13 2021-03-23 Nuvaira, Inc. Apparatus for treating asthma using neurotoxin
US9339618B2 (en) 2003-05-13 2016-05-17 Holaira, Inc. Method and apparatus for controlling narrowing of at least one airway
US8172827B2 (en) 2003-05-13 2012-05-08 Innovative Pulmonary Solutions, Inc. Apparatus for treating asthma using neurotoxin
US20060225742A1 (en) * 2003-05-13 2006-10-12 The Foundry, Inc. Apparatus for treating asthma using neurotoxin
US8073538B2 (en) 2003-11-13 2011-12-06 Cardio Polymers, Inc. Treatment of cardiac arrhythmia by modification of neuronal signaling through fat pads of the heart
US8401641B2 (en) 2003-11-13 2013-03-19 Cardiopolymers, Inc. Treatment of cardiac arrhythmia by modification of neuronal signaling through fat pads of the heart
US20080004662A1 (en) * 2003-11-13 2008-01-03 Peters Nicholas S Post-operative control of cardiac arrhythmia by modification of neuronal signaling through fat pads of the heart
US20050152923A1 (en) * 2004-01-08 2005-07-14 Brin Mitchell F. Methods for treating vascular disorders
US20060057166A1 (en) * 2004-01-08 2006-03-16 Allergan, Inc. Methods for treating Raynaud's syndrome
US6974579B2 (en) * 2004-01-08 2005-12-13 Allergan, Inc. Methods for treating vascular disorders
US7811584B2 (en) 2004-06-30 2010-10-12 Allergan, Inc. Multivalent clostridial toxins
US20090048431A1 (en) * 2004-06-30 2009-02-19 Allergan, Inc. Multivalent clostridial toxins
US20090004225A1 (en) * 2004-08-02 2009-01-01 Allergan, Inc. Toxin compounds with enhanced membrane translocation characteristics
US20060024331A1 (en) * 2004-08-02 2006-02-02 Ester Fernandez-Salas Toxin compounds with enhanced membrane translocation characteristics
EP1982997A1 (en) 2004-09-01 2008-10-22 Allergan, Inc. Degradable clostridial toxins
EP1982996A1 (en) 2004-09-01 2008-10-22 Allergan, Inc. Degradable clostridial toxins
US20110184330A1 (en) * 2004-09-18 2011-07-28 Asthmatx, Inc. Inactivation of smooth muscle tissue
US20060062808A1 (en) * 2004-09-18 2006-03-23 Asthmatx, Inc. Inactivation of smooth muscle tissue
US7906124B2 (en) * 2004-09-18 2011-03-15 Asthmatx, Inc. Inactivation of smooth muscle tissue
US8828945B2 (en) 2004-09-18 2014-09-09 Asthmatx, Inc. Inactivation of smooth muscle tissue
US20070259002A1 (en) * 2004-10-29 2007-11-08 John Batchelor Botulinum Toxin Therapy of Heart Rhythm Disorders
US10350004B2 (en) 2004-12-09 2019-07-16 Twelve, Inc. Intravascular treatment catheters
US11272982B2 (en) 2004-12-09 2022-03-15 Twelve, Inc. Intravascular treatment catheters
US20060211619A1 (en) * 2005-03-15 2006-09-21 Steward Lance E Multivalent clostridial toxin derivatives and methods of their use
US8623999B2 (en) 2005-03-15 2014-01-07 Allergan, Inc. Modified Clostridial toxins with enhanced targeting capabilities for endogenous Clostridial toxin receptor systems
US7514088B2 (en) 2005-03-15 2009-04-07 Allergan, Inc. Multivalent Clostridial toxin derivatives and methods of their use
EP2316847A2 (en) 2005-03-15 2011-05-04 Allergan, Inc. Modified clostridial toxins with enhanced targeting capabilities for endogenous clostridial toxin receptor systems
US11666526B2 (en) 2005-07-22 2023-06-06 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US10894011B2 (en) 2005-07-22 2021-01-19 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US20100087775A1 (en) * 2005-07-22 2010-04-08 The Foundy, Llc Methods and systems for toxin delivery to the nasal cavity
US10610675B2 (en) 2005-07-22 2020-04-07 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US8961391B2 (en) 2005-07-22 2015-02-24 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US11679077B2 (en) 2005-07-22 2023-06-20 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US8133497B2 (en) 2005-07-22 2012-03-13 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US8338164B2 (en) 2005-07-22 2012-12-25 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US8105817B2 (en) 2005-07-22 2012-01-31 The Foundry Llc Methods and systems for toxin delivery to the nasal cavity
US10022529B2 (en) 2005-07-22 2018-07-17 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US7655243B2 (en) 2005-07-22 2010-02-02 The Foundry, Llc Methods and systems for toxin delivery to the nasal cavity
US9498283B2 (en) 2005-07-22 2016-11-22 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US10729897B2 (en) 2005-07-22 2020-08-04 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US9700707B2 (en) 2005-07-22 2017-07-11 The Foundry, Llc Methods and systems for toxin delivery to the nasal cavity
US20070025919A1 (en) * 2005-07-22 2007-02-01 The Foundry Inc. Systems and methods for delivery of a therapeutic agent
US8636684B2 (en) 2005-07-22 2014-01-28 The Foundry Llc Methods and systems for toxin delivery to the nasal cavity
US10052465B2 (en) 2005-07-22 2018-08-21 The Foundry, Llc Methods and systems for toxin delivery to the nasal cavity
US7608275B2 (en) 2005-07-22 2009-10-27 The Foundry, Llc Systems and methods for delivery of a therapeutic agent
US7794386B2 (en) 2006-03-15 2010-09-14 Allergan, Inc. Methods for facilitating weight loss
US9511210B2 (en) 2006-05-19 2016-12-06 The Foundry, Llc Apparatus for toxin delivery to the nasal cavity
US20070267011A1 (en) * 2006-05-19 2007-11-22 The Foundry Inc. Apparatus for toxin delivery to the nasal cavity
US20080092910A1 (en) * 2006-10-18 2008-04-24 Allergan, Inc. Apparatus and method for treating obesity using neurotoxins in conjunction with bariatric procedures
US8489192B1 (en) 2008-02-15 2013-07-16 Holaira, Inc. System and method for bronchial dilation
US11058879B2 (en) 2008-02-15 2021-07-13 Nuvaira, Inc. System and method for bronchial dilation
US8483831B1 (en) 2008-02-15 2013-07-09 Holaira, Inc. System and method for bronchial dilation
US8731672B2 (en) 2008-02-15 2014-05-20 Holaira, Inc. System and method for bronchial dilation
US9125643B2 (en) 2008-02-15 2015-09-08 Holaira, Inc. System and method for bronchial dilation
US8470337B2 (en) 2008-03-13 2013-06-25 Allergan, Inc. Therapeutic treatments using botulinum neurotoxin
US20090232850A1 (en) * 2008-03-13 2009-09-17 Manack Aubrey N Therapeutic treatments using botulinum neurotoxin
US8808280B2 (en) 2008-05-09 2014-08-19 Holaira, Inc. Systems, assemblies, and methods for treating a bronchial tree
US8821489B2 (en) 2008-05-09 2014-09-02 Holaira, Inc. Systems, assemblies, and methods for treating a bronchial tree
US8088127B2 (en) 2008-05-09 2012-01-03 Innovative Pulmonary Solutions, Inc. Systems, assemblies, and methods for treating a bronchial tree
US8961507B2 (en) 2008-05-09 2015-02-24 Holaira, Inc. Systems, assemblies, and methods for treating a bronchial tree
US8226638B2 (en) 2008-05-09 2012-07-24 Innovative Pulmonary Solutions, Inc. Systems, assemblies, and methods for treating a bronchial tree
US8961508B2 (en) 2008-05-09 2015-02-24 Holaira, Inc. Systems, assemblies, and methods for treating a bronchial tree
US10149714B2 (en) 2008-05-09 2018-12-11 Nuvaira, Inc. Systems, assemblies, and methods for treating a bronchial tree
US9668809B2 (en) 2008-05-09 2017-06-06 Holaira, Inc. Systems, assemblies, and methods for treating a bronchial tree
US9675412B2 (en) 2009-10-27 2017-06-13 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US9649153B2 (en) 2009-10-27 2017-05-16 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US9005195B2 (en) 2009-10-27 2015-04-14 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US8777943B2 (en) 2009-10-27 2014-07-15 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US8932289B2 (en) 2009-10-27 2015-01-13 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US8740895B2 (en) 2009-10-27 2014-06-03 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US9931162B2 (en) 2009-10-27 2018-04-03 Nuvaira, Inc. Delivery devices with coolable energy emitting assemblies
US9017324B2 (en) 2009-10-27 2015-04-28 Holaira, Inc. Delivery devices with coolable energy emitting assemblies
US9649154B2 (en) 2009-11-11 2017-05-16 Holaira, Inc. Non-invasive and minimally invasive denervation methods and systems for performing the same
US10610283B2 (en) 2009-11-11 2020-04-07 Nuvaira, Inc. Non-invasive and minimally invasive denervation methods and systems for performing the same
US11712283B2 (en) 2009-11-11 2023-08-01 Nuvaira, Inc. Non-invasive and minimally invasive denervation methods and systems for performing the same
US11389233B2 (en) 2009-11-11 2022-07-19 Nuvaira, Inc. Systems, apparatuses, and methods for treating tissue and controlling stenosis
US9149328B2 (en) 2009-11-11 2015-10-06 Holaira, Inc. Systems, apparatuses, and methods for treating tissue and controlling stenosis
US8911439B2 (en) 2009-11-11 2014-12-16 Holaira, Inc. Non-invasive and minimally invasive denervation methods and systems for performing the same
US8768451B2 (en) 2010-04-30 2014-07-01 Boston Scientific Scimed, Inc. Therapeutic agent delivery device for delivery of a neurotoxin
US9919144B2 (en) 2011-04-08 2018-03-20 Medtronic Adrian Luxembourg S.a.r.l. Iontophoresis drug delivery system and method for denervation of the renal sympathetic nerve and iontophoretic drug delivery
WO2014053651A1 (en) 2012-10-04 2014-04-10 Dublin City University Biotherapy for pain
US9398933B2 (en) 2012-12-27 2016-07-26 Holaira, Inc. Methods for improving drug efficacy including a combination of drug administration and nerve modulation
EP3943105A2 (en) 2013-07-09 2022-01-26 Ipsen Bioinnovation Limited Cationic neurotoxins
US9920310B2 (en) 2013-07-09 2018-03-20 Ipsen Bioinnovation, Limited Cationic neurotoxins
US11034947B2 (en) 2013-07-09 2021-06-15 Ipsen Bioinnovation Limited Cationic neurotoxins
US10149893B2 (en) 2013-09-24 2018-12-11 Allergan, Inc. Methods for modifying progression of osteoarthritis
US10457927B2 (en) 2013-12-23 2019-10-29 Dublin City University Multiprotease therapeutics for chronic pain
US20170246268A1 (en) * 2014-09-12 2017-08-31 Gary E.l BORODIC Longevity Enhancement Induced by Repetitive Injections of Botulinum Toxin (Centurion Effect)
EP3822286A1 (en) 2015-01-09 2021-05-19 Ipsen Bioinnovation Limited Cationic neurotoxins
EP3650462A1 (en) 2015-10-02 2020-05-13 Ipsen Biopharm Limited Method for purifying clostridial neurotoxin
EP3889168A1 (en) 2016-05-05 2021-10-06 Ipsen Biopharm Limited Chimeric neurotoxins
WO2018002348A1 (en) 2016-07-01 2018-01-04 Ipsen Biopharm Limited Production of activated clostridial neurotoxins
WO2018060351A1 (en) 2016-09-29 2018-04-05 Ipsen Biopharm Limited Hybrid neurotoxins
US11116561B2 (en) 2018-01-24 2021-09-14 Medtronic Ardian Luxembourg S.A.R.L. Devices, agents, and associated methods for selective modulation of renal nerves
WO2019162696A1 (en) 2018-02-26 2019-08-29 Ipsen Biopharm Limited Use of ultrasound to guide injection of non-cytotoxic protease
WO2022189807A2 (en) 2021-03-11 2022-09-15 Ipsen Biopharm Limited Modified clostridial neurotoxins

Also Published As

Publication number Publication date
DE60312309T3 (en) 2011-02-24
WO2003084567A1 (en) 2003-10-16
AU2003220511A1 (en) 2003-10-20
US20030185860A1 (en) 2003-10-02
US7494654B2 (en) 2009-02-24
JP2013053167A (en) 2013-03-21
DE60312309T2 (en) 2007-11-08
AU2003220511B2 (en) 2006-11-16
KR20040105818A (en) 2004-12-16
US7048927B2 (en) 2006-05-23
DE60312309D1 (en) 2007-04-19
US20040142005A1 (en) 2004-07-22
EP1490097B2 (en) 2010-08-18
US7223399B2 (en) 2007-05-29
JP2005521735A (en) 2005-07-21
BR0308928A (en) 2005-01-04
EP1490097B1 (en) 2007-03-07
ES2281636T5 (en) 2010-12-17
US20060127417A1 (en) 2006-06-15
US20040223975A1 (en) 2004-11-11
EP1490097A1 (en) 2004-12-29
HK1070575A1 (en) 2005-06-24
MXPA04009410A (en) 2005-01-25
JP2009102440A (en) 2009-05-14
CA2480505A1 (en) 2003-10-16
ATE355852T1 (en) 2007-03-15
ES2281636T3 (en) 2007-10-01
AU2003220511C1 (en) 2008-03-06

Similar Documents

Publication Publication Date Title
US6767544B2 (en) Methods for treating cardiovascular diseases with botulinum toxin
US7671177B2 (en) Leucine-based motif and clostridial neurotoxins
AU2003243770B2 (en) Botulinum toxins for treating priapism
WO2006138059A2 (en) Treatment of autoimmune disorders with a neurotoxin
AU2007256920B2 (en) Use of a botulinum toxin for treating a urethral stricture
AU2007200658A1 (en) Use of botulinum toxin for treating cardiovascular diseases
US7419675B2 (en) Method for treating peritoneal adhesions

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALLERGAN SALES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BROOKS, GREGORY F.;REEL/FRAME:012791/0495

Effective date: 20020329

AS Assignment

Owner name: ALLERGAN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DONOVAN, STEPHEN;REEL/FRAME:013888/0200

Effective date: 20030320

AS Assignment

Owner name: ALLERGAN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ALLERGAN SALES, INC. (MERGED INTO ALLERGAN SALES, LLC 6/3/2002);REEL/FRAME:013898/0170

Effective date: 20030331

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

REMI Maintenance fee reminder mailed
FPAY Fee payment

Year of fee payment: 8

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20160727